Sie sind auf Seite 1von 15

Biochimica et Biophysica Acta 1826 (2012) 443–457

Contents lists available at SciVerse ScienceDirect

Biochimica et Biophysica Acta


journal homepage: www.elsevier.com/locate/bbacan

Review

Ascorbic acid: Chemistry, biology and the treatment of cancer☆


Juan Du a, Joseph J. Cullen a, b, c, d, Garry R. Buettner a, c,⁎
a
Department of Radiation Oncology, University of Iowa College of Medicine, Iowa City, IA, USA
b
Department of Surgery, University of Iowa College of Medicine, Iowa City, IA, USA
c
Holden Comprehensive Cancer Center, USA
d
Veterans Affairs Medical Center, Iowa City, IA, USA

a r t i c l e i n f o a b s t r a c t

Article history: Since the discovery of vitamin C, the number of its known biological functions is continually expanding. Both
Received 1 March 2012 the names ascorbic acid and vitamin C reflect its antiscorbutic properties due to its role in the synthesis of
Received in revised form 11 June 2012 collagen in connective tissues. Ascorbate acts as an electron-donor keeping iron in the ferrous state thereby
Accepted 13 June 2012
maintaining the full activity of collagen hydroxylases; parallel reactions with a variety of dioxygenases affect
Available online 20 June 2012
the expression of a wide array of genes, for example via the HIF system, as well as via the epigenetic land-
Keywords:
scape of cells and tissues. In fact, all known physiological and biochemical functions of ascorbate are due to
Ascorbate its action as an electron donor. The ability to donate one or two electrons makes AscH − an excellent reducing
Oxidative stress agent and antioxidant. Ascorbate readily undergoes pH-dependent autoxidation producing hydrogen perox-
Hydrogen peroxide ide (H2O2). In the presence of catalytic metals this oxidation is accelerated. In this review, we show that the
Cancer chemical and biochemical nature of ascorbate contribute to its antioxidant as well as its prooxidant proper-
Pharmacology ties. Recent pharmacokinetic data indicate that intravenous (i.v.) administration of ascorbate bypasses the
Nutrition tight control of the gut producing highly elevated plasma levels; ascorbate at very high levels can act as
prodrug to deliver a significant flux of H2O2 to tumors. This new knowledge has rekindled interest and
spurred new research into the clinical potential of pharmacological ascorbate. Knowledge and understanding
of the mechanisms of action of pharmacological ascorbate bring a rationale to its use to treat disease especial-
ly the use of i.v. delivery of pharmacological ascorbate as an adjuvant in the treatment of cancer.
Published by Elsevier B.V.

Contents

1. Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 444
2. Chemistry and biochemistry . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 444
3. Biosynthesis and uptake . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 444
4. Biological functions . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 446
4.1. Ascorbate as enzyme cofactors of hydroxylases . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 446
4.2. Ascorbate as a cofactor in the HIF system . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 447
4.3. Ascorbate as a cofactor in histone demethylation . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 447
4.4. Ascorbate and cancer prevention . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 447
5. Recycling of ascorbate . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 448
5.1. Reduction of ascorbate free radical . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 448
5.2. Reduction of dehydroascorbate . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 448
6. Ascorbate as an antioxidant . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 448
7. Pro-oxidant effects of ascorbate . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 448

Abbreviations: Asc, a general abbreviation to include all forms, i.e. AscH2 + AscH− + Asc2 − + DHA; AscH2, ascorbic acid, vitamin C; AscH−, ascorbate monoanion; Asc2 −, ascor-
bate dianion; Asc•−, semidehydroascorbate, i.e. ascorbate radical; DHA, dehydroascorbic acid; HIF, hypoxia inducible factor; PDI, protein disulfide isomerase; SVCT,
sodium-dependent vitamin C transporters
☆ Supported by NIH grants CA137230 and 1R01GM073929, the Medical Research Service, Department of Veterans Affairs, and the Susan L. Bader Foundation of Hope.
⁎ Corresponding author at: Department of Radiation Oncology, Free Radical and Radiation Biology, Med Labs B180, University of Iowa College of Medicine, Iowa City, IA 52242,
USA. Tel.: +1 319 335 8015 (office), +1 319 335 8019 (secretary); fax: +1 319 335 8039.
E-mail address: garry-buettner@uiowa.edu (G.R. Buettner).

0304-419X/$ – see front matter. Published by Elsevier B.V.


doi:10.1016/j.bbcan.2012.06.003
444 J. Du et al. / Biochimica et Biophysica Acta 1826 (2012) 443–457

8. Ascorbate and cancer treatment . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 449


8.1. The role of hydrogen peroxide generation and removal . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 449
8.2. Ascorbate-induced cytotoxicity in vitro . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 450
8.3. High dose ascorbate in animal studies . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 450
8.4. Clinical studies . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 452
8.5. Ascorbate and conventional cancer therapy . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 452
9. Perspectives . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 452
10. Summary . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 453
References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 453

1. Introduction Ascorbate oxidizes readily. The rate of oxidation is dependent on pH


and is accelerated by catalytic metals [14]. In the absence of catalytic
In the 80 years since the discovery of vitamin C (ascorbic acid, AscH2; metals, the spontaneous oxidation of ascorbate is quite slow at pH 7.0
ascorbate, AscH−) [1,2], the number of its known biological functions is [15]. This autoxidation, i.e. oxidation in the absence of catalytic metals,
continually expanding. Because of the ease of oxidation of ascorbate, occurs via the ascorbate dianion, Asc 2− [16]. At pH 7.0 the dominant
gaining the first understanding of its role as the antiscorbutic vitamin species for vitamin C is AscH− (99.9%) with low concentrations of
was a major challenge. As a water-soluble reducing agent and donor an- AscH2 (0.1%) and Asc 2− (0.005%). The amount of Asc 2− will increase
tioxidant, AscH− can undergo two consecutive, one-electron oxidations by a factor of ten with a one unit increase in pH; this will increase
resulting in the formation of ascorbate radical (Asc•−) and dehy- the rate of autoxidation by a factor of 10. In an air-saturated phosphate
droascorbic acid (DHA) (Fig. 1). Ascorbate can be regenerated from ascor- buffer (pH 7.0), it has been estimated that the observed pseudo-
bate radical and DHA, either enzymatically or non-enzymatically. first-order rate constant for the autoxidation of ascorbate is on the
Ascorbate readily undergoes pH-dependent autoxidation producing hy- order of 10−7–10−6 s−1 at pH 7.0 [15], consistent with a rate constant
drogen peroxide [3]. In the presence of catalytic metals this oxidation is of ≈300 M−1 s −1 for the true autoxidation of Asc2−, Reaction 2 [17].
accelerated [4]. Ascorbate can also have pro-oxidant effects. In fact the
combination of iron and ascorbate has long been used as an oxidizing sys- k ¼ 300 M−1 s−1 ð2Þ
2− •− •−
tem; the combination of these two reagents is referred to as the Asc þ O2 →Asc þ O2 :
Udenfriend system and is used for the hydroxylation of alkanes, aro-
matics, and other oxidations [5,6]. In addition, ascorbate serves as a reduc- Thus, true autoxidation is indeed very slow. In most laboratory set-
ing cofactor for many enzymes, for example hydroxylases that belong to tings, oxidation of ascorbate is catalyzed by adventitious catalytic metals
the Fe2+–2-oxoglutarate-dependent families of dioxygenases. in the buffers as well as metals that enter the buffer for laboratory equip-
The uptake of ascorbate from the intestinal tract is very tightly ment [15,18]. In near-neutral buffers with contaminating metals, the ox-
controlled [7]. However, recent pharmacokinetic data indicate that idation and subsequent loss of ascorbate can be very rapid.
intravenous administration of ascorbate can bypass this tight control
resulting in highly elevated plasma levels [8]. Because ascorbate read- 3. Biosynthesis and uptake
ily oxidizes to produce H2O2, pharmacological ascorbate has been
proposed as a prodrug for the delivery of H2O2 to tumors [9–12]. Plants and most animals synthesize ascorbate from glucose. In primi-
This new knowledge brings insights into the controversial role of tive fish, amphibians and reptiles, ascorbate synthesis takes place in kid-
ascorbate in the treatment of cancer. In this review we present the ney, whereas liver is the site of synthesis in mammals. Humans, other
fundamental chemistry and biochemistry of ascorbate that may primates, guinea-pigs and a few species of fruit-eating bats cannot syn-
have a role in the mechanisms of high dose, i.v. ascorbate in the treat- thesize ascorbate because the gene encoding L-gulonolactone oxidase
ment of cancer. (GLO), the enzyme required for the last step in ascorbate synthesis, is
not functional [19]. Thus, dietary intake becomes vital, i.e. it is a vitamin.
2. Chemistry and biochemistry The typical human diet contains both ascorbate and DHA; absorption
occurs in the enterocytes of the small intestine. While ascorbate is accu-
Ascorbic acid (AscH2, vitamin C) is a water-soluble ketolactone mulated in cells by Na+-dependent vitamin C transporters (SVCTs),
with two ionizable hydroxyl groups. It has two pKa's, pK1 is 4.2 and DHA is absorbed via a Na+-independent facilitative glucose transporters
pK2 is 11.6; thus, the ascorbate monoanion, AscH −, is the dominant (GLUTs) followed by intracellular reduction [20–24]. Under physiological
form at physiological pH (Fig. 1). Ascorbate is an excellent reducing conditions, DHA concentrations in plasma are very low, 2 μM or less,
agent and readily undergoes two consecutive, one-electron oxida- while plasma glucose levels are significantly higher (2–5 mM). Thus,
tions to form ascorbate radical (Asc •−) and dehydroascorbic acid high intracellular ascorbate concentrations are mostly due to the uptake
(DHA). The ascorbate radical is relatively unreactive due to resonance of ascorbate by SVCT1 (SLC23A1) and SVCT2 (SLC23A2) [25,26]. Human
stabilization of the unpaired electron; it readily dismutes to ascorbate SVCT1 (Km ≈65–240 μM) resides largely in brush-border surfaces of in-
and DHA (kobs = 2 × 10 5 M −1 s −1, pH 7.0) [4]. testinal and renal tubular cells to mediate absorption and re-absorption
[21]. Intestinal absorption and renal re-absorption determines the bio-
•− þ −
2Asc þ H ↔AscH þ DHA: ð1Þ availability of ascorbate. Because of the limited affinity of SVCT1 for ascor-
bate, plasma ascorbate concentrations are tightly controlled [8]. SVCT2
These properties make ascorbate an effective donor antioxidant (Km ≈8–62 μM) is found in a range of neural, neuroendocrine, exocrine,
[13]. and endothelial tissues as well as in osteoblasts [25,27]. Both isoforms of
Pure ascorbic acid is a white crystalline powder, extremely soluble SVCT are sensitive to the changes in ascorbate levels [28,29]; it has been
in water making a colorless solution. Sodium ascorbate often contains observed that high intracellular ascorbate lowered SVCTs in human plate-
significant quantities of oxidation products departing a yellow color. lets and in an intestinal epithelial cell line Caco-2TC7, while low levels
At pH values between 6 and 7.8, the purity and concentration of an up-regulated SVCTs. In addition, both SVCT1 and SVCT2 are remarkably
ascorbate solution can be easily determined by its absorbance, specific for L-ascorbate; ascorbate-2-O-phosphate, DHA, glucose, and
ε265 = 14,500 M −1 s −1 [14]. 2-deoxyglucose are not transported by SVCTs. The SVCTs have a greater
J. Du et al. / Biochimica et Biophysica Acta 1826 (2012) 443–457 445

accumulate DHA through GLUTs [30]. It has been hypothesized that the
SVCT pathway maintains intracellular levels of ascorbate under normal
physiologic conditions, whereas the GLUT-mediated DHA uptake is re-
stricted to specialized oxidizing conditions, such as those found in the sur-
rounding milieu of the activated neutrophils [30]. Given that cancer
development and progression is considered to have an inflammatory
component [31], it is highly possible that ascorbate, is oxidized in the ex-
tracellular environment, and then taken up by tumor as well as stromal
cells [32].
DHA is taken up with lower affinity (Km = 0.8 mM) than ascorbate
(Km = 0.2 mM), but the maximal rates of uptake by adult human
jejunum are similar for DHA and ascorbate (Vmax 28 vs. 35 pmol s−1
mg-protein−1) when glucose is absent [33]. Although the structure of
DHA has similarities to glucose, the influence of glucose on DHA uptake
varies between cell types. In adipocytes, neutrophils, osteoblasts and
smooth muscle cells, the uptake of DHA is largely inhibited by physio-
logical concentrations of glucose, which occurs to a lesser extent in
astrocytes, enterocytes, and renal tubular cells [27]. Mammalian facilita-
tive glucose transporters GLUT1 and GLUT3 mediate DHA transport
with a similar efficiency to that of glucose. GLUT4 transports DHA
with a higher affinity than 2-deoxyglucose (Km = 0.98 vs. 5.2 mM) but
a much lower Vmax [34]. Given that: (1) cellular uptake of ascorbate is
regulated by both glucose and insulin [35,36]; and (2) ascorbate infu-
sion has been shown to enhance glucose uptake and whole body glu-
cose disposal in healthy subjects and diabetics [37], it is clear that
more research is needed on the effects of ascorbate infusion and tissue
uptake.
Human erythrocytes express a high number of GLUT1, but have no
SVCT proteins [38]. The ascorbate level in erythrocytes is similar to the
Fig. 1. Structures of the chemical species associated with vitamin C. The structures in
blue are those forms that dominate the biochemistry of vitamin C. plasma from which they were taken [39,40]. It is speculated that erythro-
cytes recycle and release ascorbate to help maintain plasma levels of
ascorbate. With the exception of erythrocytes, intracellular ascorbate con-
affinity for L-ascorbate than D-isoascorbate; they depend on sodium to centrations are higher than extracellular fluids. Studies have shown that
create the electrochemical gradient across the plasma membrane, thereby ascorbate accumulates to millimolar concentrations in neutrophils, lym-
providing the energy required for uptake of ascorbate [27]. Recently an phocytes, monocytes, and platelets (Table 1). In circulating lymphocytes,
ascorbate analog 6-bromo-6-deoxy-L-ascorbic acid has been identified 4 mM can be achieved in healthy young women with oral ascorbate
as completely specific for SVCTs but not GLUT1 or -3, the authors demon- supplementation [41]. Intracellular ascorbate not only contributes to the
strated that 6-bromo-6-deoxy-L-ascorbic acid was taken up through intracellular reducing environment, it can also quench extracellular oxi-
the SVCTs of human skin fibroblasts but not by neutrophils, which dants by transferring electrons across the plasma membrane [42]. In

Table 1
Ascorbate content of human tissues.

Tissue Ascorbate Ascorbate Ref.


(mmol/kg wet tissue) (mM)

Adrenal glands 1.7–2.3 [45]


Pituitary gland 2.3–2.8 [45]
Liver 0.8–1 [45]
Spleen 0.8–1 [45]
Pancreas 0.8–1 [45]
Kidneys 0.28–0.85 [45]
Skeletal muscle 0.17–0.23 [45]
Brain 0.74–0.85 [45,226,227,234]
Placenta 0.23–0.72 [48]
Plasma (healthy) 0.04–0.08 [41,45,51]
Red blood cell 0.04–0.06 [39]
Neutrophil 1.2 [41]
Lymphocyte 4.0 [41]
Monocyte 3.2 [41]
Platelet 3.7 [41,56]
Cerebral spinal fluid 0.15–0.25a [235–237]
Neuron 10 [238]
Glial cells 1 [238]
Lens 2.5–3.4 [46]
Corneal epithelium 12.5 [49]
Aqueous humor 0.4–1.1 [46]
Alveolar macrophage 0.32 [50]
Bronchoalveolar lavage 0.04–0.06 [51]
Saliva 0.04–0.05 [45]
a
Ascorbate levels in cerebral spinal fluid are approximately three to four times that of plasma.
446 J. Du et al. / Biochimica et Biophysica Acta 1826 (2012) 443–457

addition, there appears to be regulated ascorbate efflux from astrocytes to H2O2 as does ascorbate. Thus, the use of ascorbate 2-phosphate is pre-
neurons, thereby bolstering antioxidant defense [43]. Furthermore, the ferred when studying the biology of vitamin C in cell culture. Millimolar
release of ascorbate from cells may also reduce non-transferrin-bound concentrations of ascorbate can be achieved intracellularly by
iron, thereby stimulating the uptake of iron by cells [44]. carrier-mediated mechanisms (Table 2). There is no detectable level
In human and animal tissues, the highest concentrations of ascorbate of DHA in cells as intracellular DHA is readily reduced by an array of bi-
are in the adrenal and pituitary glands (Table 1) [45–48]. Ascorbate is a ological reductants and enzyme systems [55,56].
physiological reductant in the dopamine β-hydroxylase-catalyzed reac- At physiological conditions DHA rapidly hydrolyzes to 2,3-L-
tion that converts dopamine to norepinephrine in the chromaffin diketogulonate (2,3-DKG), which is quite unstable [57]. 2,3-DKG subse-
granules of adrenal medulla. Likewise, peptidyl glycine α-amidating quently decarboxylates yielding L-xylonate and L-lyxonate; these com-
monooxygenase in the pituitary glands also requires high concentrations pounds can enter the pentose phosphate pathways [58]. Alternatively,
of ascorbate to catalyze the amidation reaction of carboxyl-terminal gly- L-erythrulose and oxalate can be produced from 2,3-DKG degradation. It
cine residue of a number of peptides. In addition, millimolar levels of has been proposed that highly reactive L-erythrulose rapidly glycates
ascorbate also found in eye tissues [46,49]; this may protect these tissues and crosslinks proteins. In fact, ascorbate-dependent modification of pro-
from potential damage caused by solar radiation. Since tissues from teins was proposed to occur in human lens during diabetic and
lungs are constantly exposed to relatively high concentrations of oxygen cataract formation. Oxalate monoalkylamide, one of the advanced
and ROS, antioxidant enzymes and low-molecular-weight antioxidants, glycation end products (AGEs) is a major product induced by incubation
such as ascorbate, serve to protect cells and tissues from oxidative dam- of human lens protein with DKG and other degradation product of ascor-
age. Ascorbate levels in respiratory tract lining fluids are similar or lower bate [59]. Oxalate is one of the major end products of ascorbate break-
than those in plasma [50]. However, there appears to be increased ascor- down in humans, and it has the potential to crystallize as calcium
bate content in the epithelial lining fluid and alveolar macrophage of oxalate in the urinary space in susceptible people [60,61]. However, in a
smokers who have a relatively high dietary intake of vitamin C [51]. study involving 16 cancer patients who had normal renal function, intra-
This increased content of ascorbate suggests that there is an adaptive de- venous administration of ascorbate ranging from 0.2 to 1.5 g kg−1 body
fense mechanism against the ROS derived from cigarette smoke. weight, resulted in approximately 30 to 80 mg of oxalic acid being excret-
Human cell culture experiments usually lack ascorbate, as these cells ed during the 6 h after the infusion [62]. While in primary hyperoxaluria,
are incapable of synthesizing this vitamin; in addition, there is no oxalic acid excretion ranged between 100 mg d−1 and 400 mg d−1. Ox-
source of vitamin C in typical cell culture media formulations. However, alate nephrocalcinosis and calcium oxalate stones develop over months to
cultured cells take up ascorbate, ascorbate-2-phosphate, as well as DHA years [63]. Overall, these studies suggest that patients with normal renal
when supplemented in the media. DHA is rapidly transported into cells functions and a time-limited course of ascorbate infusions would not
by glucose transporters over a period of minutes while ascorbate and its have an immediate risk of oxalate stone formation.
phosphate require SVCTs and can take several hours [52]. Ascorbate
2-phosphate is most likely hydrolyzed by membrane esterases with 4. Biological functions
the resulting ascorbate being transported into cells [53]. Compared to
the same concentration of ascorbate, cells accumulate intracellular 4.1. Ascorbate as enzyme cofactors of hydroxylases
ascorbate via ascorbate 2-phosphate at a somewhat slower rate to
reach the same intracellular concentration [54]. However, ascorbate In addition to the monooxygenases mentioned above, ascorbate is
2-phosphate does not oxidize in the media and thus does not produce involved in many physiological and biochemical processes involving

Table 2
Ascorbate in cultured cells.

Cell culture Treatment Intracellular ascorbate Ref


(mM)

A431 (human epidermoid carcinoma) 1 mM ascorbatea, 18 h 1 [239]


Adj.PC-5 mouse plasmacytoma 200 μM Ascb, 12 h 7 [240]
Alveolar macrophage (rat) 0.1 mM Asc, 30 min 3.2 [241]
Alveolar type II epithelial cells (rat) 0.1 mM Asc, 30 min 3.2 [241]
Astrocytes (rat) DHAc, 100 μM, 10 min 1.5 [248]
B lymphocytes 250 μM Asc, 4 h 0.7–1.5 [242]
EA.hy926 endothelial cells 0.3 mM Asc, 60 min 3–4 [244]
HepG2 hepatic epithelial cells 32 μM 14C-Asc, 3 min 1.1 [245]
HL-60 0.4–3 mM ascorbic phosphate 0.5 [246]
HUVEC 1 mM ascorbate, 18 h 3.5
INS-1 (rat pancreatic β-cell) 0.4 mM Asc, 16–18 h 2.4 [247]
K562 (human erythroleukemia) 500 μM DHA, 30 min 3 [248]
L6 skeletal myocytes (rat) 100 μM DHA, 10 min 12.5 [242]
d
MIA PaCa-2 pancreatic cancer 50 μM Asc, 4 h 6
1 mM Asc, 4 h 8
15 mM Asc, 4 h 18
Murine microvascular endothelial cells 0.5 mM Asc, 12 h 5 [249]
Osteoblasts (mouse) 1 mM Asc, 24 h 8
Primary hepatocytes 32 μM 14C-Asc, 3 min 0.41 [245]
SH-SY5Y neuroblastoma 2 mM Asc, 16–18 h 6 [250]
Skin fibroblasts 1 mM ascorbate, 18 h 0.4 [239]
U937 (human histocytic leukemia) 50 μM Asc or Asc-2-phosphate, 24 h 4 [54]
U937 1 mM Asc or Asc-2-phosphate, 24 h 7 [54]
a
Ascorbate.
b
L-Ascorbic acid.
c
Dehydroascorbate.
d
Thomas J van't Erve, personal communication.
J. Du et al. / Biochimica et Biophysica Acta 1826 (2012) 443–457 447

enzymatic reactions that are catalyzed by members of the Fe 2+– 300 μM [65], well below the millimolar concentrations of ascorbate
2-oxoglutarate-dependent family of dioxygenases. These dioxygen- in most of cells in healthy individuals [55]. Supplementation of ascor-
ases use 2-oxoglutarate as a co-substrate and as a source for two of bate in a lymphoma cell model in SCID mice has been shown to di-
the four electrons needed for the reduction of O2; they transfer one minish HIF levels and the rate of tumor growth [79]. Lu et al.
oxygen atom from O2 to the succinate product and one atom to the demonstrated that the antitumorigenic effect of ascorbate is depen-
protein substrate. Ascorbate is required for maintaining iron in the dent on the activity of HIF hydroxylases [80]. By analyzing endometri-
ferrous state, thereby maintaining full activity of this class of en- al tumor tissues for ascorbate and three of its targets GLUT-1, BNIP3
zymes. Reaction 3 [64,65]. and VEGF, Kuiper et al. [81] found that HIF-1α and its targets were
all up-regulated in human tumor tissue. Tumors with higher ascor-
3þ − 2þ
Fe  dioxygenase ðinactiveÞ þ AscH →Fe ð3Þ bate levels had low HIF-1 activation, while tumors with high HIF-1
•−
dioxygenase ðactiveÞ þ Asc : activation and aggressive characteristics had lower ascorbate levels.
These important findings indicate that having high tissue levels of
Evidence that the Fe 2+–2-oxoglutarate-dependent dioxygenases ascorbate may protect against HIF-1 activation and aggressive
can modify proteins was gathered from the studies of hydroxylases tumor behavior.
in collagen metabolism. Located within the lumen of the endoplasmic As a downstream factor produced upon activation of HIF, VEGF
reticulum, collagen prolyl-4-hydroxylase and prolyl-3-hydroxylase plays an important role in tumor angiogenesis. In addition to direct
are responsible for the insertion of the hydroxyl groups onto prolyl cytotoxicity effects, pharmacological ascorbate has been shown to
residues, which is essential for the proper assembly of collagen [66]. suppress capillary-like tube formation on cells grown on Matrigel
There are at least three isozymes of collagen prolyl hydroxylase in [82]. However, it is still unknown whether the suppression of angio-
humans each with distinct α subunits; but they all have protein disul- genesis by pharmacological ascorbate is due to the production of
fide isomerases (PDIs) to serve as β subunits, which catalyze the for- H2O2 [83] or through the inhibition of HIF [84].
mation of intra- and inter-chain disulfide bonds during collagen
synthesis [67]. Collagens are major constituents of the extracellular
4.3. Ascorbate as a cofactor in histone demethylation
matrix (ECM), constituting ≈30% of total protein mass. Type I colla-
gen is the major structural protein in the interstitial ECM, while
The newly discovered histone demethylases containing Jumonji cat-
type IV is prevalent in basement membrane [68]. Collagens in the ex-
alytic domains, which can demethylate trimethylated lysines, also be-
tracellular matrix are important components of the physical barrier
long to the Fe 2+–2-oxoglutarate dioxygenase family [85]. As with
against invasion and metastasis of cancer cells [88,89]. Ascorbate
other members of this family, the Jumonji proteins have the ability to
has been shown to stimulate the production of types I and III collagen
bind Fe2+, use 2-oxoglutarate as substrate, and require ascorbate for
in human skin fibroblasts [70]. Cameron and Pauling proposed that
full catalytic activity. Furthermore, ascorbate supplementation also
mega doses of ascorbate would inhibit cancer growth by preventing
leads to increased histone acetylation as well as increased expression
cancer cell invasion [71]. More recently, it has been shown that a
of HIF histone demethylases JMJD 1 and 2 in human embryonic stem
type IV collagen domain, produced as a recombinant protein, inhibits
cells [86]. Thus, we can speculate that epigenetic status of tissue may
the proliferation of capillary endothelial cells and blood vessel forma-
also depend on ascorbate to keep Fe2+–2-oxoglutarate dioxygenases
tion, resulting in the suppression of tumor growth [72]. However, it is
functional [87]. Indeed, Esteban et al. [88] also postulated that ascorbate
not known if very high levels of ascorbate will influence cancer
supplementation may enhance the function of epigenetic modifiers
growth by mechanisms related to collagen synthesis.
during reprogramming of induced pluripotent stem cells (iPSCs), as
many of these modifiers are ascorbate-dependent dioxygenases. Con-
sidering that cancer cells have shared features of normal stem cells
4.2. Ascorbate as a cofactor in the HIF system
[89] and DNA hypermethylation is associated with cancer development,
ascorbate and other nutritional supplements could prevent cancer by
As novel members of the dioxygenase family, cytoplasmic prolyl
modulating the components of the systems that set the epigenetic
hydroxylases have been shown to regulate hypoxia-inducible tran-
code [90].
scription factor (HIF) [73]. These enzymes have no PDI subunits but
The necessity of ascorbate as a reducing agent to maintain full
have higher values of Km for oxygen than collagen prolyl hydroxy-
functioning of an array of enzymes shows the importance of vitamin
lases. HIF protein is composed of an oxygen-sensitive HIF-α subunit
C in maintaining the biochemical machinery of cells and tissues.
and an oxygen-insensitive HIF-β subunit. In hypoxia, HIF-α is in-
These functions are often overlooked as many researchers focus
duced, enters the nucleus, and forms a heterodimer with HIF-β,
only on the antioxidant abilities of ascorbate.
which is constitutively expressed. In normoxia, HIF-α is efficiently
hydroxylated by prolyl hydroxylases. Hydroxylated HIF-α then
binds to the von Hippel–Lindau (pVHL) protein, which is part of an 4.4. Ascorbate and cancer prevention
ubiquitin ligase complex, leading to ubiquitination and proteasomal
degradation of the protein [74]. All three of the HIF prolyl hydroxy- Epidemiologic evidence suggests that vitamin C-rich foods play a
lases (PHD1, PHD2, and PHD3) recognize the C-terminal hydroxyl- protective role against development of cancer [91–94], plasma concen-
ation site with a higher affinity than the N-terminal hydroxylation trations of ascorbate have been shown to be inversely associated with
site [75]. In addition, an asparaginyl hydroxylase inhibits HIF through cancer risk [95,96]; however, large scale randomized intervention
the hydroxylation of a C-terminal asparagine residue, blocking trials comparing antioxidants (vitamins A, C, E and β-carotene) supple-
coactivator recruitment. Given that there are more than 800 direct ments given alone or in combination have not shown protective effects
HIF target genes yielding more than 60 gene products that are regu- [97–99]. One of the drawbacks of randomized control trials (RCT) for
lated by HIF [76–78], it is likely that the availability of ascorbate will nutrients is that most nutrients such as vitamins have threshold behav-
have a profound effect on many cell functions such as angiogenesis, ior [100]. For example, scurvy only develops when serum ascorbate
cell survival, glucose uptake, glycolysis and iron homeostasis. levels are on the order of 8 μM or less [8]. The recommended dietary al-
As members of the Fe 2+–2-oxoglutarate-dependent dioxygenase lowance (RDA) for vitamin C is 75–125 mg daily; consistent intake of
families, HIF prolyl- and asparaginyl hydroxylases require ascorbate 60 mg d−1 will in general prevent scurvy for 30–45 days once vitamin
as a reducing agent to maintain iron in the ferrous state. Values of C intake ceased [101,102]. The fact that ascorbate is required to main-
Km for ascorbate with the HIF prolyl hydroxylases range from 140 to tain full function of an array of enzymes indicates optimizing intake
448 J. Du et al. / Biochimica et Biophysica Acta 1826 (2012) 443–457

would optimize metabolism as well as prevent cancer and other degen- 6. Ascorbate as an antioxidant
erative disease [103,104].
The typical concentration of ascorbate in plasma from healthy humans
5. Recycling of ascorbate is about 40–80 μM (Table 1). At these levels ascorbate functions as an en-
dogenous antioxidant; for example, it serves as a co-antioxidant with vi-
5.1. Reduction of ascorbate free radical tamin E to protect LDL from detectable oxidative damage induced by
aqueous peroxyl radicals [115]. Thermodynamically ascorbate readily do-
There are several enzymatic systems involved in the recycling of nates an electron to potentially damaging oxidizing radicals such as hy-
ascorbate radical (Asc•−) and DHA. The reduction of ascorbate radical droxyl radical (HO•), alkoxyl radical (RO•), peroxyl radical (LOO•), thiol
by NADH-cytochrome b5 reductase has been shown in rat liver [105], radical (GS•), and tocopheroxyl radicals (TO•) [13]; this one-electron oxi-
where most of the reductase activity is localized in the outer mitochon- dation of AscH− results in the production of the ascorbate radical (Asc•−).
dria membrane. As a membrane-bound protein, NADH-cytochrome b5 Ascorbate radical is relatively unreactive, and can be reduced back to
reductase is also associated with the endoplasmic reticulum and the ascorbate by NADH- and NADPH-dependent reductases [116]; Asc•−
plasma membrane [106]. Cytochrome b5 donates an electron to ascor- can also undergo a pH-dependent disproportionation reaction, resulting
bate radical, which in turn, draws electrons from NADH. On the other in the formation ascorbate and DHA, Reaction 1 [117,118].
hand, the transmembrane ascorbate radical reductase in erythrocytes Physiological concentrations of ascorbate have been shown to
can use electrons from NADH or intracellular ascorbate to regenerate inhibit LDL oxidation induced by endothelial cells, macrophages, copper
extracellular ascorbate from ascorbate radical [107]. The plasma mem- ions, and AAPH (2,2′-azo-bis (2-amidinoproprane) dihydrochloride)
brane ascorbate radical reductase reduces ascorbate radical generated [119,120]. An important feature of the action of ascorbate is its synergistic
in blood during oxidative stress as well as the ascorbate radical generat- interaction with vitamin E [121]. Vitamin E is lipid-soluble; it is a primary
ed by reduction of α-tocoperoxyl radical during lipid oxidation. Reduc- antioxidant in LDL and lipid membrane oxidation [122]. Its one-electron
tion of ascorbate radical by thioredoxin reductase (TrxR) purified from oxidation product, the α-tocoperoxyl radical, can be reduced by ascor-
rat liver suggests that TrxR can also function as a cytosolic ascorbate bate. The two-electron oxidation product, tocopherol quinone, un-
radical reductase, which may complement cellular ascorbate recycling. dergoes ring breakage and thus vitamin E is lost [123]. In this context,
Thus, there are several biochemical routes for the one-electron reduc- ascorbate is important for maintaining vitamin E and inhibiting lipid ox-
tion of ascorbate radical to regenerate ascorbate. idation. It has been shown that doubling plasma ascorbate concentra-
tions by supplementation results in decreased rates of disappearance of
5.2. Reduction of dehydroascorbate vitamin E in smokers [124]. This is the first in vivo demonstration that
ascorbate maintains vitamin E, most likely through “recycling”.
DHA is unstable at physiological pH with a half-life is 5–15 min at
• •
37 °C [108–110]. It is estimated that the concentration of DHA is less LOO þ TOH→LOOH þ TO ð4Þ
than 2 μM in healthy humans; many reported measurements may − • •−
have overestimated DHA levels due to the ease of oxidation of AscH þ TO →Asc þ TOH: ð5Þ
AscH − during sample processing. Extracellular DHA can be imported Ascorbate is clearly a co-antioxidant protecting lipid oxidation even in
into cells using glucose transporters present in cells, where DHA can iron-loaded human plasma [125]. In human plasma lipid peroxides
be reduced to ascorbate by glutathione (GSH) directly [111] or more (cholesteryl ester hydroperoxides) were detectable only when serum
efficiently by glutaredoxin in the cytoplasm [112]. Purified rat liver ascorbate was depleted; iron-induced lipid peroxidation was inhibited
thioredoxin reductase also catalyzes NADPH-dependent reduction of by ascorbate in a concentration dependent manner [126]. Guinea pigs
DHA [113]. Thus, electrons from NADPH can find their way to DHA with iron overload had greater oxidative lipid damage that is suppressed
via thiols. with oral supplementation of ascorbate [127]. Thus, in the presence of cat-
Purified bovine liver PDI has been found to react directly with DHA alytic iron, robust nutritional levels of ascorbate protect from oxidative
and GSH to catalyze the reduction of DHA to ascorbate [112]. The damage.
finding that PDI has DHA reductase activity is interesting since
human collagen prolyl 4-hydroxylase has PDI as a β subunit [69]. It 7. Pro-oxidant effects of ascorbate
is possible that the β subunit also serves as DHA reductase to generate
ascorbate from DHA since collagen hydroxylases require ascorbate to In the presence of catalytic metal ions, ascorbate can also exert
maintain iron in its ferrous state. pro-oxidant effects [14,128,129]. Ascorbate is an excellent one-electron
3α-Hydroxysteroid dehydrogenase belongs to the family of oxido- reducing agent that can reduce ferric (Fe3+) to ferrous (Fe2+) iron,
reductases that catalyze NADPH-dependent oxidoreduction of a variety while being oxidized to ascorbate radical (Reaction 6). Depending on co-
of substrates. Del Bello et al. demonstrated that the NADPH-dependent ordination environment, Fe2+ can readily react with O2, reducing it to su-
DHA reductase from rat liver cytosol is identical to 3α-hydroxysteroid peroxide radical (Reaction 7), which in turn dismutes to H2O2 and O2
dehydrogenase [114]. The enzyme has a low affinity for DHA (Km = (Reaction 8).
4.6 mM) but a high affinity for NADPH (Km b 5 μM). Under pathological
− 3þ •− 2þ
conditions, decreases in GSH and increases in DHA could activate AscH þ Fe →Asc þ Fe ð6Þ
3α-hydroxysteroid dehydrogenase.
2þ 3þ •−
Thus, DHA has many routes for its two-electron reduction to Fe þ O2 →Fe þ O2 ð7Þ
regenerate ascorbate. Because of the relative instability of DHA, rapid
reduction to ascorbate also prevents loss of vitamin C. Ascorbate is in- •− •− þ
O2 þ O2 þ 2H →H2 O2 þ O2 : ð8Þ
volved in a variety of biochemical functions that are fundamental to
the functioning of a wide variety of enzyme systems; thus efficient In a classic Fenton reaction, Fe2+ reacts with H2O2 to generate Fe 3+
recycling maintains the pool of ascorbate in cells and tissues. and the very oxidizing hydroxyl radical, Reaction 9. The presence of
The regeneration of ascorbate from ascorbate radical and DHA will ascorbate can allow the recycling of Fe3+ back to Fe2+, which in turn
allow many cycles of ascorbate oxidation, thus one molecule of ascor- will catalyze the formation of highly reactive oxidants from H2O2.
bate can produce many molecules of H2O2. This mechanism is an im-
portant one when considering pharmacological ascorbate in cancer
2þ 3þ •
treatment that will be discussed later. Fe þ H2 O2 →Fe þ HO ðFenton reactionÞ: ð9Þ
J. Du et al. / Biochimica et Biophysica Acta 1826 (2012) 443–457 449

Depending on concentrations, the effects of ascorbate on models iron from cellular ferritin [158]. Ferritin is only one candidate as a
of lipid peroxidation can be pro- or antioxidant [14,130]. There is con- source of catalytic iron; extracellular iron chelates are present in tis-
siderable variability in the literature; this variability appears to be a sue and seem to be increased under pathological conditions [159]. It
result of the different concentrations and form of transition metal is clear that only low levels of catalytic metals are needed to substan-
ions in the experiments and the media [131]. tially increase the rate of oxidation of ascorbate [15,18]. These many
The prooxidant effects of ascorbate may be important in vivo observations provide insights on the mechanism by which pharmaco-
depending on the availability of catalytic metal ions. In healthy individ- logic concentrations of ascorbate have potential in treating certain
uals, iron is largely sequestered by iron binding proteins such as trans- types of cancer.
ferrin and ferritin [132,133]. Transferrin is a glycoprotein that is
synthesized in the liver. It is the major circulating iron binding protein
8. Ascorbate and cancer treatment
with a high affinity, but low capacity for iron; this iron is essentially
redox inactive [134]. Transferrin avidly binds to the transferrin receptor
The use of high-dose ascorbate in treating cancer patients began in
on the cell surface. The transferrin–transferrin receptor complex is in-
the 1970s. These early studies demonstrated beneficial effects of
ternalized to an endosome, which releases iron in acidic conditions.
high-dose ascorbate [155–158]. However, two double-blinded, ran-
The ferric iron released is reduced and transported to the cytoplasm
domized clinical trials at the Mayo Clinic did not show any benefit
where it is either utilized for synthesis of iron-containing proteins or
[169,160]. Subsequently, use of ascorbate for cancer treatment was
bound in ferritin, an iron storage protein. Ferritin is capable of seques-
considered ineffective and dismissed by the research and medical
tering up to 4500 atoms of iron, but is normally only 20% saturated.
communities. However, a marked difference existed in these studies.
Iron stored in ferritin can be released by appropriate reductants in the
Cameron's group gave patients ascorbate intravenously as well as
presence of a chelator or by degradation of ferritin in the lysosome.
orally, while patients in the Mayo Clinic trials received only oral
Iron can be released from ferritin by biological reductants such as
ascorbate. Some years later, clinical data were generated that demon-
thiols, ascorbate and reduced flavins [135]. The released iron enables
strated that when ascorbate is given orally, plasma concentrations are
cells to synthesize cytochromes and iron-containing enzymes. How-
tightly controlled [101]. At oral doses of 200 mg, the steady-state
ever, uncontrolled release of iron from ferritin has the potential to
plasma concentrations are ≈80 μM. As doses exceed 200 mg, relative
form HO •, which can damage critical cellular components [136–138].
absorption decreases, urine excretion increases and the fraction of
In pathological situations, such as thalassaemia or hemotochro-
bioavailable ascorbate is reduced [7]. Peak plasma values do not ex-
matosis, non-transferrin-bound iron is present. Thus, supplemental
ceed ≈220 μM even after maximum oral dose of 3 g 6 times daily
ascorbate without administration of an iron chelator can lead to delete-
[8]. In contrast, when ascorbate is administered intravenously, milli-
rious effects [14]. Tissue damage resulting from ischemia/reperfusion is
molar concentrations can be achieved. In fact, infusion of 10 g of
another example of increased availability of catalytic metal occurring in
ascorbate in cancer patients results in plasma concentrations of 1 to
vivo [139]. Intravenous ascorbate prior to vascular surgery increased
5 mM [161,162]. Thus, only intravenous administration of ascorbate
concentrations of ascorbate radical and lipid hydroperoxides suggesting
can yield high plasma levels, i.e. pharmacological levels.
that catalytic iron released into the circulation during the ischemic
phase of the surgery with ascorbate may promote iron-induced lipid
peroxidation [140,141]. 8.1. The role of hydrogen peroxide generation and removal
Elevated levels of catalytic metal ions have also been demonstrat-
ed in chronic inflammatory diseases [142]. There is an increased de- In vitro, the toxicity of ascorbate centers on the generation of H2O2
position of iron proteins in the synovial membranes in rheumatoid by ascorbate upon its oxidation [9,10,12,163,164]. The true autoxida-
arthritis. Ascorbate radical has been detected in synovial fluid from tion of ascorbate, i.e. in the absence of catalytic metals, via reaction 2
patients with synovitis disease indicating that catalytic iron is in will generate considerable amounts of H2O2 when ascorbate is at mil-
part responsible for the decreased levels of ascorbate and increased limolar levels. For example, in an aqueous solution containing 20 mM
levels of DHA [141]. In addition, ascorbate concentrations were de- ascorbate at pH 7.4, the concentration of ascorbate dianion, Asc 2−,
creased while levels of catalytic iron increased in patients with sepsis, will be on the order of 1 μM. This will result in a flux of H2O2 on the
compared to healthy subjects [143]. order of 10 nM s −1, in a typical cell culture experiment.
Although there is no direct evidence that catalytic iron is increased Catalytic iron (and perhaps copper) in the media and serum will
in tumors, many patients with malignant disease have elevated increase the rate of ascorbate oxidation and associated generation of
serum or tissue ferritin concentrations [144–146]. Raised levels of cir- H2O2. Clement et al. found that Dulbecco's modification of Eagle's
culating ferritin are found in childhood Hodgkins lymphoma and are MEM (DMEM) generates more H2O2 than RPMI 1640 during a
associated with poor survival [147]; serum ferritin levels have been 6-hour incubation with increasing concentrations of ascorbate [165].
shown to be related to the stage of the disease and tumor volume in In addition to adventitious iron, DMEM has 0.25 μM Fe (NO3)3 in its
cervical cancer [148]. Furthermore, studies by Feng et al. [149] have formulation, which could contribute to this greater production of
shown that the peritoneal and subcutaneous microvessels of normal H2O2 in DMEM than that from RPMI. Serum usually contains total
mice were largely impermeable to circulating ferritin, but in mice iron at a range of 10 to 50 μM, which could also be another factor
bearing solid or ascites tumors, circulating ferritin was found in the that causes the variable toxicity of ascorbate in different media; how-
basal lamina and extravascular space, suggesting that tumor vessels ever, some of this iron will be redox inactive as it will be sequestered
are hyperpermeable to circulating macromolecules such as ferritin. in transferrin [134]; there will undoubtedly be a highly variable
In fact, ferritin staining was detected in stroma and histiocytes sur- amount of iron present that has nonspecific peroxidase activity
rounding neoplastic cells of breast carcinoma tissue of patients [166]. This iron will actively catalyze the oxidation of ascorbate.
[150]. Extracellular metal-containing proteins have been proposed Thus, the amount of catalytic iron present in typical cell culture
to be essential for the pro-oxidant effects of ascorbate [10,129], media is highly variable. Even with careful attention to detail, this
iron-saturated ferritin could be a potential candidate as source of cat- can lead to considerable variability in experimental results, even in
alytic iron. A recent study by Deubzer et al. [151] demonstrated that the same set of experiments.
ferritin released by neuroblastoma cells enhanced pharmacologic α-Keto acids such as pyruvate and α-ketoglutarate directly react
ascorbate induced-cytotoxicity, indicating that ferritin with high with H2O2 when present in cell culture media [172];
iron-saturation could be a source of catalytic iron [152]. Consistent
with this, ascorbate has also been shown to be capable of releasing CH3 ð¼ OÞð¼ OÞOH þ H2 O2 →CH3 Cð¼ OÞOH þ CO2 ð10Þ
450 J. Du et al. / Biochimica et Biophysica Acta 1826 (2012) 443–457

this will result in an apparent lowering of the toxicity of ascorbate reactions with components of the media. The key is that H2O2 is re-
[168]. Thus, the presence of α-keto acids in media must be taken moved. Thus, interpretation of data from experiments with iron,
into consideration in the design and interpretation of data from ex- ascorbate, H2O2 or some combination is not always straightforward.
periments where the oxidation of ascorbate and associated produc- Oxidative stress has been shown to increase the levels of catalytic
tion of H2O2 are central to the study. iron in tissues. A series of electron paramagnetic resonance (EPR)
Because cells rapidly and efficiently remove extracellular H2O2, ob- studies demonstrated that ultraviolet light increased the levels of la-
served toxicities or cell killing induced by ascorbate is an inverse func- bile iron in skin [186]; heat stress-induced oxidative events increased
tion of cell density [151,153,169]. There is an array of intracellular the level of labile iron in liver [187]; and ischemia reperfusion in-
antioxidant enzymes involved in the removal of H2O2, such as catalase, creased the level of catalytic iron in the heart [188]. In addition, ion-
glutathione peroxidase, and the peroxiredoxins. Although modest cata- izing radiation and some chemotherapeutic drugs have been shown
lase activity has been detected in heart mitochondria [170], catalase is to increase catalytic free iron levels [189–191]. Since it takes only
predominantly located in peroxisomes of mammalian cells. Concomi- very low concentrations of catalytic metals to bring about the rapid
tant administration of the catalase inhibitor amino-1,2,4-triazole has oxidation of ascorbate [192,193], approaches that increase catalytic
been shown to enhance ascorbate toxicity to Ehrlich ascites cells in iron could potentially enhance the cytotoxicity of pharmacologic
vitro [171]. Using adenovirus constructs containing human catalase ascorbate in vivo [194].
cDNA, Du et al. [12] demonstrated that overexpressing intracellular cat- Porphyrin-based SOD mimics have a redox-active metal (Mn, Fe,
alase protected cells from ascorbate-induced cytotoxicity. These results and Cu) center and a stable porphyrin complex. The dismutation of
provide evidence supporting a fundamental role for H2O2 in O2•− by Mn porphyrin complexes involves two steps in which the Mn
ascorbate-induced toxicity. center cycles between Mn(III) and Mn(II): the first step is the reduc-
Cytosolic glutathione peroxidase (GPx1), another antioxidant en- tion of Mn(III) by O2•− to yield Mn(II) and O2 and the second step the
zyme that removes peroxide, is widely distributed in tissues. GPx1 oxidation of Mn(II) by O2•− to yield H2O2 and return the manganese
catalyzes GSH-dependent reduction of H2O2 to water. Gaetani et al. to its resting state as Mn(III) porphyrin [195]. However, in the pres-
[172,173] have shown that the intracellular concentration of H2O2 ence of a reductant such as ascorbate, Mn porphyrins function as su-
in human erythrocytes is inversely proportional to the activity of cat- peroxide reductases rather than dismutases; Mn(III) can be reduced
alase and GPx-1. GPx1 is responsible for eliminating low concentra- to Mn(II) by ascorbate while Mn(II) can react with O2 forming O2•−,
tions of H2O2; however, at high fluxes of H2O2 recycling of GPx1 by which subsequently forms H2O2 and O2, Reaction 8. The prooxidant ef-
GSH becomes rate-limiting [174]; at high fluxes of H2O2 catalase is fects of Mn porphyrin and ascorbate have been studied by several
the enzyme that is principally responsible for removal of H2O2 [175]. groups. Gardner et al. [196] have demonstrated that MnTM-4-PyP 5+
Another antioxidant system that contributes to removal of H2O2 catalyzes the oxidation of ascorbate in vitro. Zhong et al. [197] have
within cells is the family of peroxiredoxins, in particular, peroxiredoxin shown synergistic killing of human prostate cancer cell RWPE-2 by
2 (Prx2). Prx2 is the third most abundant protein in erythrocytes MnTM-4-PyP 5+ and ascorbate. Most recently, Tian et al. [198] have
[176]. The rate constant of human Prx2 reacting with H2O2 is shown that MnTM-4-PyP 5+ synergizes with ascorbate inhibiting the
1.3 × 10 7 M − 1 s − 1 [177] which is similar to that of catalase (1.7 × growth of prostatic, pancreatic, and hepatic cancer cells. Furthermore,
10 7 M − 1 s − 1 [178]). These complementary enzymes systems for the cytotoxic effects of two Mn(III) alkylpyridylporphyrins (MnTE-2-
the removal of intracellular H2O2 in RBCs will also make these cells PyP 5+ and MnTnHex-2-PyP 5+) and ascorbate have been demonstrat-
efficient sinks for H2O2. ed in Caco-2, HeLa, HCT116, and 4T1 cells [199,200]. The more lipo-
philic MnTnHex-2-PyP 5+ appeared much more effective. Given that
8.2. Ascorbate-induced cytotoxicity in vitro several Mn porphyrins have already been tested in vivo as SOD mi-
metics, and by themselves have shown low toxicities at micromolar
Chen et al. [9,11] have demonstrated that some cancer cells have in- levels [195], there is great potential for using Mn porphyrins as an ad-
creased sensitivity to ascorbate-induced cytotoxicity compared to nor- juvant to enhance the efficacy of pharmacologic ascorbate.
mal cells. In a complementary study, Du et al. [12] demonstrated that
pancreatic cancer cells are more sensitive to pharmacological concen- 8.3. High dose ascorbate in animal studies
trations of ascorbate than their normal cell counterparts. The difference
in sensitivity between normal and cancer cells towards ascorbate may As mentioned above, the uptake of oral ascorbate in humans is
be due to low levels of antioxidant enzymes and high endogenous levels tightly controlled by the gut and kidney filtration [7]. Rats receiving
of ROS in cancer cells [179–181]. The relative lower activities of catalase, ascorbate by gavage (0.5 mg g −1) increased blood and extracellular
glutathione peroxidase, and peroxiredoxins in cancer cells could poten- concentration to peak levels b 150 μM [10]. By contrast, concentra-
tially contribute to less efficient removal of H2O2 and increased sensitiv- tions reached peak levels of nearly 3 mM in rats receiving intraperito-
ity to ascorbate-induced cytotoxicity. neal injections, while intravenous injection increased peak levels to
The rate of oxidation of ascorbate is typically a function of the level >8 mM. In a similar study, mice receiving bolus intravenous injec-
of catalytically active iron and copper in solution. Iron in cell culture tions of ascorbate (1 g kg −1), resulted in plasma concentrations of
media contributes significantly to the rate of H2O2 generation. 15 mM [164]. Supplementation of ascorbate in drinking water at
Deferoxamine (Desferal® or DFO) is an iron-chelating agent that ren- 1 g kg −1 only increased plasma concentrations to b 50 μM. These re-
ders iron catalytically inactive with respect to ascorbate oxidation sults clearly indicate that pharmacologic concentrations of ascorbate
[15]. Although hydrophilic, DFO is cell-permeable; short-term expo- cannot be obtained by oral administration.
sure to DFO is not effective in accessing cytosolic labile iron pool In addition to the finding that millimolar levels of ascorbate were
(LIP) of cells from different origins [182]. However, short-term achieved by parenteral administration, Chen and colleagues [10]
pre-incubation of cells to DFO appeared to protect cells from demonstrated that ascorbate radical was formed in extracellular
ascorbate-induced toxicity [184]. These observations indicate that fluid but was not detectable in whole blood. Moreover, H2O2 was
DFO either is effective in accessing endosomal iron [182] or iron asso- detected in extracellular fluid, but not in the blood; H2O2 correlated
ciated with cellular membranes [183]. Other cell culture studies have with ascorbate radical concentration. These results indicate that plas-
demonstrated that Fe 2+ in the media can actually protect cells from ma membrane associated ascorbate radical reductase and high levels
the oxidative damage resulting from exposure to extracellular H2O2 of catalase, glutathione peroxidase, and peroxiredoxin enable eryth-
[184,185]. Although hydroxyl radicals are produced via the Fenton re- rocytes to act as a sink for extracellular ascorbate radical and H2O2
action, most will not induce cell damage, but rather disappear by [176,107]. On the other hand, ascorbate could be oxidized by catalytic
J. Du et al. / Biochimica et Biophysica Acta 1826 (2012) 443–457 451

iron associated with damaged proteins in the extracellular space the extracellular space could accumulate to a concentration greater
[201–203, 209]. Considering that the permeability of tumor vessels is than the intracellular level resulting in net rate of diffusion across the
much higher compared to normal endothelium, tumor endothelium cell membrane into cells, resulting in toxicity to cancer cells [10].
may permit the outflow of macromolecules, such as albumin, to intersti- Pharmacological ascorbate inhibits tumor growth in mice. In mice
tial fluid [169,204]. The fluid that accumulated in the peritoneal bearing tumor xenografts of pancreatic cancer, treatment with
cavities of ascites tumor-bearing mice had a protein concentration 4 g kg −1 ascorbate (i.p., twice daily) significantly decreased the rate
several-fold greater than that of normal peritoneal fluid, with approxi- of tumor growth [11,12,164] (Table 4). Upon cessation of treatment
mately 85% of the protein level of plasma, including albumin, in a pro- with pharmacological ascorbate, the rate of tumor growth increased.
portion similar to that found in plasma [205,206]. Human albumin is This rate was similar to the rate observed in the control group, i.e. no
the most abundant protein in the plasma; and has the capacity to bind ascorbate (Du et al., unpublished results). In this tumor model phar-
to metal ions such as Cu2+ [207]. The first four amino acids of the macologic ascorbate as a single agent was cytostatic, not cytotoxic. Ob-
N-terminus of human albumin Asp-Ala-His-Lys forms a tight-binding viously, the use of intravenous high dose ascorbate as a single agent for
site for Cu2+ [207]. In fact, almost thirty years ago, Linus Pauling and cancer was not curative. To test the combination effect of standard che-
his colleagues [194] designed a copper:glycylglycylhistidine complex motherapy with ascorbate, Espey et al. [209] have shown that synergis-
that mimics the binding of albumin to copper; it enhanced the antitumor tic cytotoxic effects can be achieved with gemcitabine and ascorbate in
activity of ascorbate against Ehrlich ascites tumor cells. In addition, ex- pancreatic cancer both in vitro and in a nude-mouse model. These data
tracellular fluids contain little or no catalase activity, and low levels of provide support for investigating the use of pharmacologic ascorbate as
SOD and GPx [208]. Thus, the H2O2 generated by ascorbate oxidation in an adjuvant for conventional cancer chemotherapies.

Table 3
Pro-oxidant effects of ascorbate in cultured cells.

Cell line Treatment Effects Ref

Primary human diploid fibroblasts GM5399 20–500 μM Asc Inhibit growth, DNA damage [251]
Normal human skin fibroblast CCD-25 SK Sodium ascorbate, 0.56–2.8 mM No effect [210]
Normal human colon fibroblast CCD-18 Co Sodium ascorbate, 0.1–0.4 mM No effect [210]
Human embryonic fibroblast Sodium ascorbate, 0.2 mM Inhibit growth [252]
HRE human normal renal epithelial cells Asc 1.2 mM Promote growth [253]
Human normal myeloid cells L-Ascorbic acid, 0.3 mM No effect [254]
Primary chicken embryo fibroblast Sodium ascorbate, 0.2 mM Inhibit growth [252]
AN3 CA endometrial adenocarcinoma Sodium ascorbate, 0.56–2.8 mM Inhibit growth [210]
Ehrlich ascites carcinoma Ascorbate + 3-amino-triazole Synergistic killing [171]
5637 human bladder cancer Asc EC50 b 5 mM Inhibit growth [11]
T24 human bladder cancer Asc + vitK3 Synergistic, [255]
Autoschizis,
Necrosis, apoptosis
MB231 human breast cancer 5 mM Asc Decrease clonogenic survival [9]
MCF-7 human breast cancer 5 mM Asc Decrease clonogenic survival [9]
MCF-7 human breast cancer Asc + porphyrin Apoptosis, cell cycle disruption [256]
Hs587t human breast cancer 5 mM Asc Decrease clonogenic survival [9]
HeLa human cervical cancer Sodium ascorbate, 0.25 mM; No effect [11,257]
Asc EC50 > 5 mM
HeLa human cervical cancer Asc + MGd (Motexafin gadolinium) Apoptosis, [257]
Lysosomal rupture
Normal human colon fibroblast CCD-18-Co Sodium ascorbate, 0.1–0.8 mM No effect [210]
Human colon carcinoma cells Sodium ascorbate, 0.1–3.2 mM Inhibit growth [210]
HEp-2 human epidermoid larynx carcinoma Asc 500 μM + 25 μM vitB12b Apoptosis, DNA damage [258]
A549 human lung cancer Asc EC50 b 5 mM Inhibit growth [11]
HepG2 human hepatocellular carcinoma Asc, Asc + MnTMPyP Decrease clonogenic survival, mitochondria damage. [198]
DU 145 human prostate cancer cells Sodium ascorbate, 1–10 mM; Inhibit growth [198]
Asc + MnTMPyP
LNCaP human prostate cancer cells Sodium ascorbate, 1–10 mM; Inhibit growth [198]
Asc + MnTMPyP
PC-3 human prostate cancer Sodium ascorbate, 0.56–2.3 mM; Inhibit growth [198]
Asc + MnTMPyP
MIA PaCa-2 human pancreatic carcinoma Sodium ascorbate, 0.56–2.8 mM; Inhibit growth [210]
MIA PaCa-2 human pancreatic carcinoma Asc 1–5 mM Cytotoxic, autophagy [12]
PANC-1 Asc, Asc + MnTMPyP Decrease clonogenic survival [198]
Pan02 mouse pancreatic cancer Asc 2.5–10 mM cytotoxic [11]
Human acute leukemic cells L-Ascorbic acid, 0.3 mM Inhibit growth [254]
U937 human histocytic leukemia cells L-Ascorbic acid, 50–300 μM Inhibit growth [163]
Human chronic lymphocytic leukemia Asc 0.3–5 mM; Cytotoxic [259]
Asc + ATO (Arsenic trioxide)
Kelly human neuroblastoma 0.6–5 mM Cytotoxic [151]
SK-N-SH human neuroblastoma 0.6–5 mM Cytotoxic [151]
Mouse neuroblastoma Sodium ascorbate + 5-FU Inhibit growth [222]
Sodium ascorbate + X-irradiation
9 L rat glioblastoma Asc 2.5–10 mM Cytotoxic [11]
LN18 human glioblastoma cell lines, Asc + γ-irradiation Double-stranded DNA breaks, G2/M arrest block [224]
human primary glioblastoma cells,
GL261 mouse astrocytoma cell line
B16F10 murine melanoma Asc 0.05–0.2 mM Cell cycle arrest [260]
Chinese hamster ovary (CHO) cells Ascorbate + misonidazole Inhibit growth [261]
452 J. Du et al. / Biochimica et Biophysica Acta 1826 (2012) 443–457

8.4. Clinical studies demonstrated that high-dose intravenous ascorbate was well tolerat-
ed in cancer patients with normal renal function; the combination of
Although the two clinical trials using oral ascorbate at the Mayo ascorbate infusion with standard of care chemotherapies did not in-
Clinic failed to show any benefit, numerous case reports have provided crease toxicity.
encouraging results with intravenous ascorbate therapy [162,210,211].
However, most of the cases were reported without sufficient detail 8.5. Ascorbate and conventional cancer therapy
or follow-up for evaluation. Padayatty et al. [212] examined three
well-documented cases in accordance with National Cancer Institute The cytotoxicity of high-dose intravenous ascorbate is dependent
(NCI) Best Case Series guidelines. These cases suggest that high-dose, on the ascorbate-induced production of H2O2. Hydrogen peroxide
intravenous ascorbate therapy prolonged the survival of the patients can deplete intracellular GSH, causing oxidative stress. In high doses
with advanced cancer. ascorbate acts as a pro-oxidant to kill cancer cells. Initial concerns
Intravenous ascorbate therapy is likely to be safe in most patients ex- that ascorbate might reduce the effectiveness of standard chemother-
cept under certain conditions [213]. Because the removal of H2O2 gener- apy and/or radiation therapy have not been demonstrated. Numerous
ated in blood requires glutathione, which in turn requires NADPH reports suggest that pharmacological ascorbate may actually increase
generated by G6PD, the rate-limiting enzyme of the pentose phosphate the efficacy of several chemotherapeutic drugs and radiation in vitro
pathway [214], high-dose ascorbate can induce intravascular hemolysis [164,222–224] (Table 3). Espey et al. [209] demonstrated that phar-
in patients with glucose-6-phosphate dehydrogenase (G6PD) deficiency macologic concentrations of ascorbate with gemcitabine resulted in
[215–217]. One end-product of ascorbate oxidation is oxalic acid, which a synergistic cytotoxic response in pancreatic tumor cell lines;
has the potential to crystallize as calcium oxalate in the urinary space in gemcitabine–ascorbate combinations administrated to mice bearing
patients with pre-existing renal insufficiency [218]. In addition, rare pancreatic tumor xenografts consistently enhanced inhibition of
cases of massive tumor hemorrhage have been reported in patients growth compared to gemcitabine alone. Case reports [161,212] indi-
with advanced cancer following high-dose intravenous ascorbate cate that intravenous ascorbate can be safely used with chemothera-
[155,219]. Thus, as always, caution is needed, but there are clear indica- py and radiotherapy and could potentially enhance the effects of
tions for a potential role for ascorbate in cancer treatment. conventional cancer therapy. Furthermore, intravenous ascorbate
Three phase I clinical trials of intravenous ascorbate in patients has been shown to improve the quality of life in breast cancer pa-
with advanced cancer measured plasma concentrations of ascorbate tients during chemo-/radiotherapy and aftercare [225]. Clinical trials
and evaluated clinical consequences [162,220,221]. In the study by using combinations of chemotherapy and intravenous ascorbate are
Riordan and colleagues, patients were given continuous infusion of underway at several institutions (www.clinicaltrials.gov).
0.15 to 0.7 g kg −1 day −1 for up to eight weeks; in the Hoffer study,
ascorbate was administrated three times per week at fixed doses of 9. Perspectives
0.4, 0.6, 0.9 and 1.5 g kg −1; and in the most recent study by Monti
et al. [221], patients received 50, 75 and 100 g per infusion (three in- The inhibition effects of pharmacologic ascorbate on tumor growth
fusions per week) for 8 weeks with concomitant i.v. gemcitabine and have been confirmed in many laboratories (Table 4). In murine models
oral erlotinib. In the Riordan study, ascorbate concentrations in serum elevated ascorbate levels in plasma were verified following i.p. adminis-
during therapy ranged from 0.28 to 3.8 mM. In the Hoffer study, peak tration [11,164]. It is still unknown whether cells and tissues levels are
plasma ascorbate concentrations ranged from 2.4 to 26 mM. When also elevated [226,227]. Human platelets supplemented with 500 μM
1.5 g kg −1 was administered, plasma ascorbate concentrations Asc for 30 min showed a decrease in SVCT2 levels, while in Asc depleted
exceeded 10 mM for ≈ 4.5 h [220], levels that have been shown to in- platelets SVCT2 expression was higher than native ones [29]. Thus in
duce cell killing in a variety of cancer cells [9,11,12,164]. Finally in the vivo studies for possible changes in SVCT expression levels following
Monti study, patients that received 100 g of ascorbate achieved peak ascorbate infusion are needed. Another issue is a possible transient
plasma concentrations between 25 and 32 mM. All three trials withdrawal effect upon cessation of treatment with pharmacologic

Table 4
Pro-oxidant effects of ascorbate in animal models.

Species Cell type Treatment Effects Ref

Balb/c nude mice HT29 colon cancer Asc 15 mg, 100, 1000 mg/kg, i.p., daily. 4 weeks. 7/7 mice survived at 1000 mg/kg. No carcinogenic [262]
invasion. Tumor volumes decrease. ARSs and EiFs
genes down regulated.
Balb/c nude mice K562 leukemia Vit K3 10 mg/kg, i.p.; Asc + vit K3 decrease tumor growth. [263]
Asc 1 g/kg, i.p.
Ncr nude mice Ovcar5 ovarian cancer Asc 4 g/kg, i.p., twice daily. 30 days. No adverse effects. Decrease tumor growth. [11]
Ncr nude mice Pan02 pancreatic cancer Asc 4 g/kg, i.p., daily. 14 days. No adverse effects. Decrease tumor growth. [11]
Ncr nude mice 9 L rat glioblastoma Asc 4 g/kg, i.p., twice daily. 12 days. No adverse effects. Decrease tumor growth. [11]
Prevent metastases.
Ncr nude mice Pan02 pancreatic cancer Asc 4 g/kg, i.p., daily; No side effects except osmotic stress. [209]
Gemcitabine 30, 60 mg/kg, i.p., every 4 days. 21 days. Asc + Gem significantly
decrease tumor growth.
Ncr nude mice PANC-1 pancreatic cancer Asc 4 g/kg, i.p., daily; No side effects except osmotic stress. [209]
Gemcitabine 30, 60 mg/kg, every 4 days. 33 days. Asc + Gem significantly
decrease tumor growth.
Nude mice MIA PaCa-2 pancreatic cancer Asc 4 g/kg, i.p., twice daily. 14 days. No adverse effects. Decrease tumor growth. [12]
NMRI mice TLT Murine hepatoma Asc 1 g/kg, i.p., daily. 30 days. Decrease tumor growth. [164]
Nude mice H322 non-small cell lung cancer Asc 250 mg/kg, i.p.; Asc synergistic with 101 F6 decrease tumor growth. [264]
101 F6 nanoparticle i.v.
SCID mice EHMS-10 mesothelioma cells Asc 0.88, 8.8 g/mouse, i.v. single dose. Decrease tumor growth. [265]
Balb/c mice Murine sarcoma S180 cells Asc 5.5, 30 mg/mouse, i.p., every two days. Decrease tumor growth; inhibit bFGF, VEGF [266]
and MMP2 genes.
Balb/c mice Murine sarcoma S180 cells Asc 1.5 mg/g, i.p., every three days. Inhibit tumor establishment; RKIP and annexin [267]
A5 levels increase.
J. Du et al. / Biochimica et Biophysica Acta 1826 (2012) 443–457 453

ascorbate [228]. Male guinea pigs that received ascorbate 1 g kg−1 [8] S.J. Padayatty, H. Sun, Y. Wang, H.D. Riordan, S.M. Hewitt, A. Katz, R.A. Wesley,
M. Levine, Vitamin C pharmacokinetics: implications for oral and intravenous
body weight per day by i.p. for 4 weeks had elevated plasma and uri- use, Ann. Intern. Med. 140 (2004) 533–537.
nary levels; however, in the weeks after the treatment withdrawn, [9] Q. Chen, M.G. Espey, M.C. Krishna, J.B. Mitchell, C.P. Corpe, G.R. Buettner, E.
mean plasma and urinary ascorbate were significantly lower than nor- Shacter, M. Levine, Pharmacologic ascorbic acid concentrations selectively kill
cancer cells: action as a pro-drug to deliver hydrogen peroxide to tissues, Proc.
mal [229]. It is unknown if a similar rebound effect would occur in Natl. Acad. Sci. U. S. A. 102 (2005) 13604–13609.
human subjects following high doses i.v. ascorbate. F2-isoprostane is a [10] Q. Chen, M.G. Espey, A. Sun, J. Lee, M.C. Krishna, E. Shacter, P.L. Choyke, C.
biomarker for lipid peroxidation in vivo, and its quantification in plasma Pooput, K.L. Kirk, G.R. Buettner, M. Levine, Ascorbate in pharmacologic
concentrations selectively generates ascorbate radical and hydrogen peroxide
and urine has emerged as the most reliable method to assess systemic in extracellular fluid in vivo, Proc. Natl. Acad. Sci. U. S. A. 104 (2007) 8749–8754.
oxidative stress in humans and animals [230–232]. For healthy young [11] Q. Chen, M.G. Espey, A.Y. Sun, C. Pooput, K.L. Kirk, M.C. Krishna, D.B. Khosh, J.
women taking oral doses of ascorbate 30–2500 mg daily, plasma and Drisko, M. Levine, Pharmacologic doses of ascorbate act as a prooxidant and de-
crease growth of aggressive tumor xenografts in mice, Proc. Natl. Acad. Sci. U. S. A.
urine F2-isoprostanes were unchanged [41]. It is unclear whether
105 (2008) 11105–11109.
plasma F2-isoprostanes will change following pharmacologic ascorbate [12] J. Du, S.M. Martin, M. Levine, B.A. Wagner, G.R. Buettner, S.H. Wang, A.F.
infusion in cancer patients. Furthermore, since pharmacological ascor- Taghiyev, C. Du, C.M. Knudson, J.J. Cullen, Mechanisms of ascorbate-induced
bate is a prodrug for H2O2 generation, there should be many applica- cytotoxicity in pancreatic cancer, Clin. Cancer Res. 16 (2010) 509–520.
[13] G.R. Buettner, The pecking order of free radicals and antioxidants: lipid peroxi-
tions where delivery of H2O2 via pharmacological ascorbate may have dation, [alpha]-tocopherol, and ascorbate, Arch. Biochem. Biophys. 300 (1993)
clinical benefit, such as infectious diseases caused by viruses, bacteria, 535–543.
and other pathogens [10,233]. [14] G.R. Buettner, B.A. Jurkiewicz, Catalytic metals, ascorbate and free radicals:
combinations to avoid, Radiat. Res. 145 (1996) 532–541.
[15] G.R. Buettner, In the absence of catalytic metals ascorbate does not autoxidize at
pH 7: ascorbate as a test for catalytic metals, J. Biochem. Biophys. Methods 16
10. Summary (1988) 27–40.
[16] Y. Song, G.R. Buettner, Thermodynamic and kinetic considerations for the reac-
More than eighty years since its discovery, our understanding of tion of semiquinone radicals to form superoxide and hydrogen peroxide, Free
Radic. Biol. Med. 49 (2010) 919–962.
the functions of ascorbic acid has evolved from the prevention of [17] N.H. Williams, J.K. Yandell, Outer-sphere electron-transfer reactions of ascorbate
scurvy to its potential use as a therapeutic drug for cancer treatment. anions, Aust. J. Chem. 35 (1982) 1133–1144.
Ascorbate maintains Fe 2+ of collagen hydroxylases in an active state; [18] G.R. Buettner, Ascorbate oxidation: UV absorbance of ascorbate and ESR spec-
troscopy of the ascorbyl radical as assays for iron, Free Radic. Res. Commun.
therefore it plays a pivotal role in collagen synthesis; parallel reac- 10 (1990) 5–9.
tions with a variety of dioxygenases affect the expression of a wide [19] M. Nishikimi, R. Fukuyama, S. Minoshima, N. Shimizu, K. Yagi, Cloning and chromo-
array of genes, for example via the HIF system, and possibly the epige- somal mapping of the human nonfunctional gene for L-gulono-gamma-lactone ox-
idase, the enzyme for L-ascorbic acid biosynthesis missing in man, J. Biol. Chem. 269
netic landscape of cells and tissues. The ability to donate one or two
(1994) 13685–13688.
electrons makes ascorbate an excellent reducing agent and antioxi- [20] J.C. Vera, C.I. Rivas, F.V. Velasquez, R.H. Zhang, I.I. Concha, D.W. Golde, Resolution
dant. However, in the presence of catalytic metals, ascorbate also of the facilitated transport of dehydroascorbic acid from its intracellular accu-
has pro-oxidant effects, where the redox-active metal is reduced by mulation as ascorbic acid, J. Biol. Chem. 270 (1995) 23706–23712.
[21] I. Savini, A. Rossi, C. Pierro, L. Avigliano, M.V. Catani, SVCT1 and SVCT2: key
ascorbate and then in turn reacts with oxygen, producing superoxide proteins for vitamin C uptake, Amino Acids 34 (2008) 347–355.
that subsequently dismutes to produce H2O2. [22] R.W. Welch, Y. Wang, A.J. Crossman, J.B. Park, K.L. Kirk, M. Levine, Accumulation
Apart from its biochemical functions that are met by healthy nutri- of vitamin C (ascorbate) and its oxidized metabolite dehydroascorbic acid
occurs by separate mechanisms, J. Biol. Chem. 270 (1995) 12584–12592.
tional levels, recent pharmacokinetic data indicate that intravenous ad- [23] E.J. Diliberto, G.D. Heckman, A.J. Daniels, Characterization of ascorbic acid trans-
ministration of ascorbate bypasses the tight control of the gut and renal port by adrenomedullary chromaffin cells. Evidence for Na+-dependent
excretion; thus, intravenous administration of ascorbate will produce co-transport, J. Biol. Chem. 258 (1983) 12886–12894.
[24] R.W. Welch, P. Bergsten, J.D. Butler, M. Levine, Ascorbic acid accumulation and
highly elevated plasma levels; this ascorbate will autoxidize resulting transport in human fibroblasts, Biochem. J. 294 (1993) 505–510.
in a high flux of extracellular H2O2. This H2O2 will readily diffuse into [25] H. Tsukaguchi, T. Tokui, B. Mackenzie, U.V. Berger, X.-Z. Chen, Y. Wang, R.F.
cells challenging the intracellular peroxide-removal system, initiating Brubaker, M.A. Hediger, A family of mammalian Na+-dependent L-ascorbic
acid transporters, Nature 399 (1999) 70–75.
oxidative cascades. These high fluxes of H2O2 appear to have little effect [26] C.P. Corpe, H. Tu, P. Eck, J. Wang, R. Faulhaber-Walter, J. Schnermann, S.
on normal cells but can be detrimental to certain tumor cells. Knowl- Margolis, S. Padayatty, H. Sun, Y. Wang, R.L. Nussbaum, M.G. Espey, M. Levine,
edge and understanding of these mechanisms brings a rationale to the Vitamin C transporter Slc23a1 links renal reabsorption, vitamin C tissue accu-
mulation, and perinatal survival in mice, J. Clin. Invest. 120 (2010) 1069–1083.
use of high-dose ascorbate to treat disease and thereby is reviving inter-
[27] J.X. Wilson, Regulation of vitamin C transport, Annu. Rev. Nutr. 25 (2005) 105–125.
est in the use of i.v. ascorbate in cancer treatment. The full potential of [28] I. Savini, M.V. Catani, R. Arnone, A. Rossi, G. Frega, D. Del Principe, L. Avigliano,
pharmacological ascorbate in cancer treatment will only be realized Translational control of the ascorbic acid transporter SVCT2 in human platelets,
with greater understanding of its basic mechanism of action in conjunc- Free Radic. Biol. Med. 42 (2007) 608–616.
[29] L. MacDonald, A.E. Thumser, P. Sharp, Decreased expression of the vitamin C
tion with the appropriate design of clinical trials. transporter SVCT1 by ascorbic acid in a human intestinal epithelial cell line,
Br. J. Nutr. 87 (2002) 97–100.
[30] C.P. Corpe, J.-H. Lee, O. Kwon, P. Eck, J. Narayanan, K.L. Kirk, M. Levine, 6-Bromo-
References 6-deoxy-L-ascorbic acid: an ascorbate analog specific for Na+-dependent vitamin
C transporter but not glucose transporter pathways, J. Biol. Chem. 280 (2005)
[1] J.L. Svirbely, A. Szent-Györgyi, The chemical nature of vitamin C, Biochem. J. 27 5211–5220.
(1933) 279–285. [31] M. Schafer, S. Werner, Cancer as an overhealing wound: an old hypothesis
[2] G.R. Buettner, F.Q. Schafer, Albert Szent-Györgyi: vitamin C identification, revisited, Nat. Rev. Mol. Cell Biol. 9 (2008) 628–638.
Biochemist 28 (2006) 31–33. [32] D.B. Agus, J.C. Vera, D.W. Golde, Stromal cell oxidation: a mechanism by which
[3] G. Calcutt, The formation of hydrogen peroxide during the autoxidation of tumors obtain vitamin C, Cancer Res. 59 (1999) 4555–4558.
ascorbic acid, Experientia 7 (1951) 26. [33] C. Malo, J.X. Wilson, Glucose modulates vitamin C transport in adult human
[4] B.H.J. Bielski, Chemistry of ascorbic acid radicals, In: in: P.A. Seib, B.M. Tolbert small intestinal brush border membrane vesicles, J. Nutr. 130 (2000) 63–69.
(Eds.), Ascorbic Acid: Chemistry, Metabolism, and Uses, Vol. 200, American [34] S.C. Rumsey, R. Daruwala, H. Al-Hasani, M.J. Zarnowski, I.A. Simpson, M. Levine,
Chemical Society, Washington D.C., 1982, pp. 81–100. Dehydroascorbic acid transport by GLUT4 in Xenopus oocytes and isolated rat
[5] M.M. Khan, A.E. Martell, Metal ion and metal chelate catalyzed oxidation of adipocytes, J. Biol. Chem. 275 (2000) 28246–28253.
ascorbic acid by molecular oxygen. II. Cupric and ferric chelate catalyzed oxida- [35] J.J. Cunningham, The glucose/insulin system and vitamin C: implications in
tion, J. Am. Chem. Soc. 89 (1967) 7104–7111. insulin-dependent diabetes mellitus, J. Am. Coll. Nutr. 17 (1998) 105–108.
[6] S. Udenfriend, C.T. Clark, J. Axelrod, B.B. Brodie, Ascorbic acid in aromatic [36] S. Sherry, E.P. Ralli, Further studies of the effects of insulin on the metabolism of
hydroxylation. I. A model system for aromatic hydroxylation, J. Biol. Chem. vitamin C, J. Clin. Invest. 27 (1948) 217–225.
208 (1954) 731–739. [37] G. Paolisso, A. D'Amore, V. Balbi, C. Volpe, D. Galzerano, D. Giugliano, S.
[7] J.F. Graumlich, T.M. Ludden, C. Conry-Cantilena, L.R.J. Cantilena, Y. Wang, M. Sgambato, M. Varricchio, F. D'Onofrio, Plasma vitamin C affects glucose homeo-
Levine, Pharmacokinetic model of ascorbic acid in healthy male volunteers stasis in healthy subjects and in non-insulin-dependent diabetics, Am. J. Physiol.
during depletion and repletion, Pharm. Res. 14 (1997) 1133–1139. Endocrinol. Metab. 266 (1994) E261–E268.
454 J. Du et al. / Biochimica et Biophysica Acta 1826 (2012) 443–457

[38] J.M. May, Z.-C. Qu, H. Qiao, M.J. Kouryab, Maturational loss of the vitamin C [74] P.J. Kallio, W.J. Wilson, S. O'Brien, Y. Makino, L. Poellinger, Regulation of the
transporter in erythrocytes, Biochem. Biophys. Res. Commun. 360 (2007) hypoxia-inducible transcription factor 1alpha by the ubiquitin–proteasome
295–298. pathway, J. Biol. Chem. 274 (1999) 6519–6525.
[39] R.M. Evans, L. Currie, A. Campbell, The distribution of ascorbic acid between [75] M. Hirsila, P. Koivunen, V. Gunzler, K.I. Kivirikko, J. Myllyharju, Characterization
various cellular components of blood, in normal individuals, and its relation to of the human prolyl 4-hydroxylases that modify the hypoxia-inducible factor,
the plasma concentration, Br. J. Nutr. 47 (1982) 473–482. J. Biol. Chem. 278 (2003) 30772–30780.
[40] H. Li, H. Tu, Y. Wang, M. Levine, Vitamin C in mouse and human red blood cells: [76] G.L. Semenza, Targeting HIF-1 for cancer therapy, Nat. Rev. Cancer 3 (2003)
an HPLC assay, Anal. Biochem. 426 (2012) 109–117. 721–732.
[41] M. Levine, Y. Wang, S.J. Padayatty, J. Morrow, A new recommended dietary [77] D.R. Mole, C. Blancher, R.R. Copley, P.J. Pollard, J.M. Gleadle, J. Ragoussis, P.J.
allowance of vitamin C for healthy young women, Proc. Natl. Acad. Sci. U. S. A. Ratcliffe, Genome-wide association of hypoxia-inducible factor (HIF)-1α and
98 (2001) 9842–9846. HIF-2α DNA binding with expression profiling of hypoxia-inducible transcripts,
[42] D.J.R. Lane, A. Lawen, Ascorbate and plasma membrane electron transport — J. Biol. Chem. 284 (2009) 16767–16775.
enzymes vs efflux, Free Radic. Biol. Med. 47 (2009) 485–495. [78] G.L. Semenza, Hypoxia-inducible factors: mediators of cancer progression and
[43] J.X. Wilson, Antioxidant defense of the brain: a role for astrocytes, Can. J. Physiol. targets for cancer therapy, Trends Pharmacol. Sci. 33 (2012) 207–214.
Pharmacol. 75 (1997) 1149–1163. [79] P. Gao, H. Zhang, R. Dinavahi, F. Li, Y. Xiang, V. Raman, Z.M. Bhujwalla, D.W.
[44] O. Han, M.L. Failla, A.D. Hill, E.R. Morris, J.C.J. Smith, Reduction of Fe(III) is required Felsher, L. Cheng, J. Pevsner, L.A. Lee, G.L. Semenza, C.V. Dang, HIF-dependent
for uptake of nonheme iron by Caco-2 cells, J. Nutr. 125 (1995) 1291–1299. antitumorigenic effect of antioxidants in vivo, Cancer Cell 12 (2007) 230–238.
[45] D. Hornig, Distribution of ascorbic acid, metabolites and analogues in man and [80] H. Lu, C.L. Dalgard, A. Mohyeldin, T. McFate, A.S. Tait, A. Verma, Reversible inac-
animals, Ann. N. Y. Acad. Sci. 258 (1975) 103–118. tivation of HIF-1 prolyl hydroxylases allows cell metabolism to control basal
[46] H.L. Kern, S.L. Zolot, Transport of vitamin C in the lens, Curr. Eye Res. 6 (1987) HIF-1, J. Biol. Chem. 280 (2005) 41928–41939.
885–896. [81] C. Kuiper, I.G. Molenaar, G.U. Dachs, M.J. Currie, P.H. Sykes, M.C. Vissers, Low
[47] S.C. Rumsey, M. Levine, Absorption, transport, and disposition of ascorbic acid in ascorbate levels are associated with increased hypoxia-inducible factor-1 activ-
humans, J. Nutr. Biochem. 9 (1998) 116–130. ity and an aggressive tumor phenotype in endometrial cancer, Cancer Res. 70
[48] M.L. Streeter, P. Rosso, Transport mechanisms for ascorbic acid in the human (2010) 5749–5758.
placenta, Am. J. Clin. Nutr. 34 (1981) 1706–1711. [82] N. Mikirova, T. Ichim, N. Riordan, Anti-angiogenic effect of high doses of ascorbic
[49] R.F. Brubaker, W.M. Bourne, L.A. Bachman, J.W. McLaren, Ascorbic acid con- acid, J .Transl. Med. 6 (2008) 50.
tent of human corneal epithelium, Invest. Ophthalmol. Vis. Sci. 41 (2000) [83] S.S. Pathi, P. Lei, S. Sreevalsan, G. Chadalapaka, I. Jutooru, S. Safe, Pharmacologic
1681–1683. doses of ascorbic acid repress specificity protein (Sp) transcription factors
[50] A. Van der Vliet, C.A. O'Neill, C.E. Cross, J.M. Koostra, W.G. Valz, B. Halliwell, S. and Sp-regulated genes in colon cancer cells, Nutr. Cancer 63 (2011)
Louie, Determination of low-molecular-mass antioxidant concentrations in 1133–1142.
human respiratory tract lining fluids, Am. J. Physiol. Lung Cell Mol. Physiol. [84] E.L. Pagé, D.A. Chan, A.J. Giaccia, M. Levine, D.E. Richard, Hypoxia-inducible
276 (1999) L289–L296. factor-1α stabilization in nonhypoxic conditions: role of oxidation and intracel-
[51] M.H. Bui, A. Sauty, F. Collet, P. Leuenberger, Dietary vitamin C intake and concen- lular ascorbate depletion, Mol. Biol. Cell 19 (2008) 86–94.
trations in the body fluids and cells of male smokers and nonsmokers, J. Nutr. [85] P.A. Cloos, J. Christensen, K. Agger, K. Helin, Erasing the methyl mark: histone
122 (1992) 312–316. demethylases at the center of cellular differentiation and disease, Genes Dev.
[52] J.M. May, Z. Qu, X. Li, Ascorbic acid blunts oxidant stress due to menadione in en- 22 (2008) 1115–1140.
dothelial cells, Arch. Biochem. Biophys. 411 (2003) 136–144. [86] T.-L. Chung, R.M. Brena, G. Kolle, S.M. Grimmond, B.P. Berman, P.W. Laird, M.F. Pera,
[53] T. Chepda, M. Cadau, P.H. Girin, J. Frey, A. Chamson, Monitoring of ascorbate at a E.J. Wolvetang, Vitamin C promotes widespread yet specific DNA demethylation of
constant rate in cell culture: effect on cell growth, In Vitro Cell. Dev. Biol. Anim. the epigenome in human embryonic stem cells, Stem Cells 28 (2010) 1848–1855.
37 (2001) 26–30. [87] A.R. Cyr, F.E. Domann, The redox basis of epigenetic modifications: from mech-
[54] J.M. Vislisel, F.Q. Schafer, G.R. Buettner, A simple and sensitive assay for ascor- anisms to functional consequences, Antioxid. Redox Signal. 15 (2011) 551–589.
bate using a plate reader, Anal. Biochem. 365 (2007) 31–39. [88] M.A. Esteban, D. Pei, Vitamin C improves the quality of somatic cell
[55] P.W. Washko, Y. Wang, M. Levine, Ascorbic acid recycling in human neutrophils, reprogramming, Nat. Genet. 44 (2012) 366–367.
J. Biol. Chem. 268 (1993) 15531–15535. [89] M. Widschwendter, H. Fiegl, D. Egle, E. Mueller-Holzner, G. Spizzo, C. Marth, D.J.
[56] J.V. Lloyd, P.S. Davis, H. Emery, H. Lander, Platelet ascorbic acid levels in normal Weisenberger, M. Campan, J. Young, I. Jacobs, P.W. Laird, Epigenetic stem cell
subjects and disease, J. Clin. Pathol. 25 (1972) 478–483. signature in cancer, Nat. Genet. 39 (2007) 157–158.
[57] G.L.W. Simpson, B.J. Ortwerth, The non-oxidative degradiation of ascorbic acid [90] M. Verma, Cancer control and prevention by nutrition and epigenetic
at physiological conditions, Biochim. Biophys. Acta 1501 (2000) 12–24. approaches, Antioxid. Redox Signal. 17 (2012) 355–364.
[58] G. Bánhegyi, L. Braun, M. Csala, F. Puskás, J. Mandl, Ascorbate metabolism and its [91] G. Block, Vitamin C and cancer prevention: the epidemiologic evidence, Am. J.
regulation in animals, Free Radic. Biol. Med. 23 (1997) 793–803. Clin. Nutr. 53 (1991) 270S–282S.
[59] R.H. Nagaraj, F.A. Shamsi, B. Huber, M. Pischetsrieder, Immunochemical detec- [92] C.M. Loria, M.J. Klag, L.E. Caulfield, P.K. Whelton, Vitamin C status and mortality
tion of oxalate monokylamide, an ascorbate-derived Maillard reaction product in US adults, Am. J. Clin. Nutr. 72 (2000) 139–145.
in the human lens, FEBS J. 453 (1999) 327–330. [93] R.E. Patterson, E. White, A.R. Kristal, M.L. Neuhouser, J.D. Potter, Vitamin supple-
[60] W. Chai, M. Liebman, S. Kynast-Gales, L. Massey, Oxalate absorption and endog- ments and cancer risk: the epidemiologic evidence, Cancer Causes Control
enous oxalate synthesis from ascorbate in calcium oxalate stone formers and 8 (1997) 786–802.
non-stone formers, Am. J. Kidney Dis. 44 (2004) 1060–1069. [94] K.F. Gey, Vitamins E plus C and interacting conutrients required for optimal
[61] O. Traxer, B. Huet, J. Poindexter, C.Y.C. Pak, M.S. Pearle, Effect of ascorbic acid health. A critical and constructive review of epidemiology and supplementation
consumption on urinary stone risk factors, J. Urol. 170 (2003) 397–401. data regarding cardiovascular disease and cancer, Biofactors 7 (1998) 113–174.
[62] L. Robitaille, O.A. Mamer, W.H.J. Miller, M. Levine, S. Assouline, D. Melnychuk, C. [95] C.A. Gonzalez, E. Riboli, Diet and cancer prevention: contributions from the Eu-
Rousseau, L.J. Hoffer, Oxalic acid excretion after intravenous ascorbic acid ropean Prospective Investigation into Cancer and Nutrition (EPIC) study, Euro. J.
administration, Metabolism 58 (2009) 263–269. Cancer 46 (2010) 2555–2562.
[63] J.R. Asplin, Hyperoxaluric calcium nephrolithiasis, Endocrinol. Metab. Clin. North [96] F. Musil, Z. Zadák, D. Solichová, R. Hyšpler, M. Kaška, L. Sobotka, J. Manák,
Am. 31 (2002) 927–949. Dynamics of antioxidants in patients with acute pancreatitis and in patients
[64] S. Englard, S. Seifter, The biochemical functions of ascorbic acid, Annu. Rev. Nutr. operated for colorectal cancer: a clinical study, Nutrition 21 (2005) 118–124.
6 (1986) 365–406. [97] G. Bjelakovic, D. Nikolova, R.G. Simonetti, C. Gluud, Antioxidant supplements for
[65] A. Ozer, R.K. Bruick, Non-heme dioxygenases: cellular sensors and regulators prevention of gastrointestinal cancers: a systematic review and meta-analysis,
jelly rolled into one? Nat. Chem. Biol. 3 (2007) 144–153. Lancet 364 (2004) 1219–1228.
[66] K.I. Kivirikko, D.J. Prockop, Enzymatic hydroxylation of proline and lysine in [98] J.M. Gaziano, R.J. Glynn, W.G. Christen, T. Kurth, C. Belanger, J. MacFadyen, V.
protocollagen, Proc. Natl. Acad. Sci. U. S. A. 57 (1967) 782–789. Bubes, J.E. Manson, H.D. Sesso, J.E. Buring, Vitamins E and C in the prevention
[67] J. Myllyharju, K.I. Kivirikko, Collagens, modifying enzymes and their mutations of prostate and total cancer in men: the Physicians' Health Study II randomized
in humans, flies and worms, Trends Genet. 20 (2004) 33–43. controlled trial, JAMA 301 (2009) 52–62.
[68] M. Egeblad, M.G. Rasch, V.M. Weaver, Dynamic interplay between the collagen [99] J. Lin, N.R. Cook, C. Albert, E. Zaharris, J.M. Gaziano, M. Van Denburgh, J.E. Buring,
scaffold and tumor evolution, Curr. Opin. Cell Biol. 22 (2010) 697–706. J.E. Manson, Vitamins C and E and beta carotene supplementation and cancer
[69] L.A. Liotta, Tumor invasion and metastases—role of the extracellular matrix: risk: a randomized controlled trial, J. Natl. Cancer Inst. 101 (2009) 14–23.
Rhoads Memorial Award lecture, Cancer Res. 46 (1986) 1–7. [100] R.P. Heaney, Nutrition, chronic disease, and the problem of proof, Am. J. Clin.
[70] J.C. Geesin, D. Darr, R. Kaufman, S. Murad, S.R. Pinnell, Ascorbic acid specifically Nutr. 84 (2006) 471–472.
increases type I and type III procollagen messenger RNA levels in human skin [101] M. Levine, C. Conry-Cantilena, Y. Wang, R.W. Welch, P.W. Washko, K.R.
fibroblasts, J. Investig. Dermatol. 90 (1988) 420–424. Dhariwal, J.B. Park, A. Lazarev, J. Graumlich, J. King, L.R. Cantilena, Vitamin C
[71] E. Cameron, L. Pauling, B. Leibovitz, Ascorbic acid and cancer: a review, Cancer pharmacokinetics in healthy volunteers: evidence for a recommended dietary
Res. 39 (1979) 663–681. allowance, Proc. Natl. Acad. Sci. U. S. A. 93 (1996) 3704–3709.
[72] Y. Maeshima, P.C. Colorado, A. Torre, K.A. Holthaus, J.A. Grunkemeyer, M.B. [102] B.J. Burri, R.A. Jacob, Human metabolism and the requirement for vitamin C, In:
Ericksen, H. Hopfer, Y. Xiao, I.E. Stillman, R. Kalluri, Distinct antitumor properties in: L. Packer, J. Fuchs (Eds.), Vitamin C in Health and Disease, Marcel Dekker,
of a type IV collagen domain derived from basement membrane, J. Biol. Chem. New York, 1997, pp. 341–366.
275 (2000) 21340–21348. [103] B.N. Ames, Low micronutrient intake may accelerate the degenerative diseases
[73] R.K. Bruick, S.L. McKnight, A conserved family of prolyl-4-hydroxylases that of aging through allocation of scarce micronutrients by triage, Proc. Natl. Acad.
modify HIF, Science 294 (2001) 1337–1340. Sci. U. S. A. 103 (2006) 17589–17594.
J. Du et al. / Biochimica et Biophysica Acta 1826 (2012) 443–457 455

[104] M. Levine, P. Eck, Vitamin C: working on the x-axis, Am. J. Clin. Nutr. 90 (2009) [136] R.F. Boyer, C.J. MacCleary, Superoxide ion as a primary reductant in ascorbate-
1121–1123. mediated ferritin iron release, Free Radic. Biol. Med. 3 (1987) 389–395.
[105] A. Ito, S.-i. Hayashi, T. Yoshida, Participation of a cytochrome b5-like hemoprotein of [137] K. Keyer, J.A. Imlay, Superoxide accelerates DNA damage by elevating free-iron
outer mitochondrial membrane (OM cytochrome b) in NADH-semidehydroascorbic levels, Proc. Natl. Acad. Sci. U. S. A. 93 (1996) 13635–13640.
acid reductase activity of rat liver, Biochem. Biophys. Res. Commun. 101 (1981) [138] P. Biemond, H.G. van Eijk, A.J. Swaak, J.F. Koster, Iron mobilization from ferritin
591–598. by superoxide derived from stimulated polymorphonuclear leukocytes. Possible
[106] N. Borgese, G. Pietrini, Distribution of the integral membrane protein mechanism in inflammation diseases, J. Clin. Invest. 73 (1984) 1576–1579.
NADH-cytochrome b5 reductase in rat liver cells, studied with a quantitative [139] C. Coudray, S. Pucheu, F. Boucher, J. Arnaud, J. de Leiris, A. Favier, Effect of
radioimmunoblotting assay, Biochem. J. 239 (1986) 393–403. ischemia/reperfusion sequence on cytosolic iron status and its release in the
[107] J.M. May, Z. Qu, C.E. Cobb, Recycling of the ascorbate free radical by human coronary effluent in isolated rat hearts, Biol. Trace Elem. Res. 41 (1994) 69–75.
erythrocyte membranes, Free Radic. Biol. Med. 31 (2001) 117–124. [140] D.M. Bailey, S. Raman, J. McEneny, I.S. Young, K.L. Parham, D.A. Hullin, B. Davies,
[108] A.M. Bode, L. Cunningham, R.C. Rose, Spontaneous decay of oxidized ascorbic G. McKeeman, J.M. McCord, M.H. Lewis, Vitamin C prophylaxis promotes oxida-
acid (dehydro-L-ascorbic acid) evaluated by high-pressure liquid chromatogra- tive lipid damage during surgical ischemia–reperfusion, Free Radic. Biol. Med. 40
phy, Clin. Chem. 36 (1990) 1807–1809. (2006) 591–600.
[109] H. Borsook, H.W. Davenport, C.E.P. Jeffreys, R.C. Warner, The oxidation of ascorbic [141] G.R. Buettner, W. Chamulitrat, The catalytic activity of iron in synovial fluid as
acid and its reduction in vitro and in vivo, J. Biol. Chem. 117 (1937) 237–279. monitored by the ascorbate free radical, Free Radic. Biol. Med. 8 (1990) 55–56.
[110] K. Bogdanski, H. Bogdanska, Sur la stabilite de l'acide L-dehdroascorbique dans [142] A.J. Dabbagh, C.W. Trenam, C.J. Morris, D.R. Blake, Iron in joint inflammation,
les solutions cbeaqueses, Bull. Acad. Pol. Sci. 3 (1955) 41–44. Ann. Rheum. Dis. 52 (1993) 67–73.
[111] B.S. Winkler, S.M. Orselli, T.S. Rex, The redox couple between glutathione and [143] H.F. Galley, M.J. Davies, N.R. Webster, Ascorbyl radical formation in patients with
ascorbic acid: a chemical and physiological perspective, Free Radic. Biol. Med. sepsis: effect of ascorbate loading, Free Radic. Biol. Med. 20 (1996) 139–143.
17 (1994) 333–349. [144] G. Güner, G. Kirkali, Ç. Yenisey, İ.R. Töre, Cytosol and serum ferritin in breast car-
[112] W.W. Wells, D.P. Xu, Y.F. Yang, P.A. Rocque, Mammalian thioltransferase cinoma, Cancer Lett. 67 (1992) 103–112.
(glutaredoxin) and protein disulfide isomerase have dehydroascorbate reduc- [145] Y. Miyata, S. Koga, M. Nishikido, T. Hayashi, H. Kanetake, Relationship between
tase activity, J. Biol. Chem. 265 (1990) 15361–15364. serum ferritin levels and tumour status in patients with renal cell carcinoma,
[113] J.M. May, S. Mendiratta, K.E. Hill, R.F. Burk, Reduction of dehydroascorbate to BJU Int. 88 (2001) 974–977.
ascorbate by the selenoenzyme thioredoxin reductase, J. Biol. Chem. 272 [146] D. Basso, C. Fabris, G. Del Pavero, T. Meggiato, M.P. Panozzo, D. Vianello, M.
(1997) 22607–22610. Plebani, R. Maccarato, Hepatic changes and serum ferritin in pancreatic cancer
[114] B. Del Bello, E. Maellaro, L. Sugherini, A. Santucci, M. Comporti, A.F. Casini, Puri- and other gastrointestinal diseases: the role of cholestasis, Ann. Clin. Biochem.
fication of NADPH-dependent dehydroascorbate reductase from rat liver and its 28 (1991) 34–38.
identification with 3 alpha-hydroxysteroid dehydrogenase, Biochem. J. 304 [147] H.W. Hann, B. Lange, M.W. Stahlhut, K.A. McGlynn, Prognostic importance of
(1994) 385–390. serum transferrin and ferritin in childhood Hodgkin's disease, Cancer 66
[115] B. Frei, L. England, B.N. Ames, Ascorbate is an outstanding antioxidant in human (1990) 313–316.
blood plasma, Proc. Natl. Acad. Sci. U. S. A. 86 (1989) 6377–6381. [148] A. Jacobs, Serum ferritin and malignant tumours, Med. Oncol. Tumor
[116] C.L. Linster, E. Van Schaftingen, Vitamin C. Biosynthesis, recycling and degrada- Pharmacother. 1 (1984) 149–156.
tion in mammals, FEBS J. 274 (2007) 1–22. [149] D. Feng, J.A. Nagy, A.M. Dvorak, H.F. Dvorak, Different pathways of macromole-
[117] A. Corti, A.F. Casini, A. Pompella, Cellular pathways for transport and efflux of cule extravasation from hyperpermeable tumor vessels, Microvasc. Res. 59
ascorbate and dehydroascorbate, Arch. Biochem. Biophys. 500 (2010) 107–115. (2000) 24–37.
[118] B.H.J. Bielski, A.O. Allen, H.A. Schwart, Mechanism of disproportionation of [150] R. Rossiello, M.V. Carriero, G.G. Giordano, Distribution of ferritin, transferrin and
ascorbate radicals, J. Am. Chem. Soc. 103 (1981) 3516–3518. lactoferrin in breast carcinoma tissue, J. Clin. Pathol. 37 (1984) 51–55.
[119] E. Niki, N. Noguchi, Protection of human low-density lipoprotein from oxidative [151] B. Deubzer, F. Mayer, Z. Kuci, M. Niewisch, G. Merkel, R. Handgretinger, G.
modification by vitamin C, In: in: L. Packer, J. Fuchs (Eds.), Vitamin C in Health Bruchelt, H2O2-mediated cytotoxicity of pharmacologic ascorbate concentra-
and Disease, Marcel Dekker, New York, 1997, pp. 183–192. tions to neuroblastoma cells: potential role of lactate and ferritin, Cell. Physiol.
[120] S.M. Lynch, J.D. Morrow, L.J. Roberts II, B. Frei, Formation of non-cyclooxygenase- Biochem. 25 (2010) 767–774.
derived prostanoids (F2-isoprostanes) in plasma and low density lipoprotein exposed [152] I. Rousseau, S. Puntarulo, Ferritin-dependent radical generation in rat liver
to oxidative stress in vitro, J. Clin. Invest. 93 (1994) 998–1004. homogenates, Toxicology 264 (2009) 155–161.
[121] C. Golumbic, H.A. Mattill, Antioxidants and the autoxidation of fats. XIII. The [153] S.L. Baader, E. Bill, A.X. Trautwein, G. Bruchelt, B.F. Matzanke, Mobilization of
antioxygenic action of ascorbic acid in association with tocopherols, hydroqui- iron from cellular ferritin by ascorbic acid in neuroblastoma SK-N-SH cells: an
nones and related compounds, J. Am. Chem. Soc. 63 (1941) 1279–1280. EPR study, FEBS Lett. 381 (1996) 131–134.
[122] G.W. Burton, K.U. Ingold, Vitamin E: application of the principles of physical [154] O. Olakanmi, J.B. Stokes, B.E. Britigan, Acquisition of iron bound to low molecular
organic chemistry to the exploration of its structure and function, Acc. Chem. weight chelates by human monocyte-derived macrophages, J. Immunol. 153
Res. 19 (1986) 194–201. (1994) 2691–2703.
[123] A.S. Csallany, H.H. Draper, S.N. Shah, Conversion of D-alpha-tocopherol-C14 to [155] E. Cameron, A. Campbell, The orthomolecular treatment of cancer. II. Clinical
tocopheryl-p-quinone in vivo, Arch. Biochem. Biophys. 98 (1962) 142–145. trial of high-dose ascorbic acid supplements in advanced human cancer,
[124] R.S. Bruno, S.W. Leonard, J. Atkinson, T.J. Montine, R. Ramakrishnan, T.M. Bray, Chem. Biol. Interact. 9 (1974) 285–315.
M.G. Traber, Faster plasma vitamin E disappearance in smokers is normalized [156] E. Cameron, A. Campbell, T. Jack, The orthomolecular treatment of cancer. III.
by vitamin C supplementation, Free Radic. Biol. Med. 40 (2006) 689–697. Reticulum cell sarcoma: double complete regression induced by high-dose
[125] T.M. Berger, M.C. Polidori, A. Dabbagh, P.J. Evans, B. Halliwell, J.D. Morrow, L.J. ascorbic acid therapy, Chem. Biol. Interact. 11 (1975) 387–393.
Roberts II, B. Frei, Antioxidant activity of vitamin C in iron-overloaded human [157] E. Cameron, L. Pauling, Supplemental ascorbate in the supportive treatment of
plasma, J. Biol. Chem. 272 (1997) 15656–15660. cancer: prolongation of survival times in terminal human cancer, Proc. Natl.
[126] J. Suh, B.-Z. Zhu, B. Frei, Ascorbate does not act as a pro-oxidant towards lipids Acad. Sci. U. S. A. 73 (1976) 3685–3689.
and proteins in human plasma exposed to redox-active transition metal ions [158] E. Cameron, L. Pauling, Supplemental ascorbate in the supportive treatment of
and hydrogen peroxide, Free Radic. Biol. Med. 34 (2003) 1306–1314. cancer: reevaluation of prolongation of survival times in terminal human cancer,
[127] K. Chen, J. Suh, A.C. Carr, J.D. Morrow, J. Zeind, B. Frei, Vitamin C suppresses ox- Proc. Natl. Acad. Sci. U. S. A. 75 (1978) 4538–4542.
idative lipid damage in vivo, even in the presence of iron overload, Am. J. Physiol. [159] E.T. Creagan, C.G. Moertel, J.R. O'Fallon, A.J. Schutt, M.J. O'Connell, J. Rubin, S.
Endocrinol. Metab. 279 (2000) E1406–E1412. Frytak, Failure of high-dose vitamin C (ascorbic acid) therapy to benefit patients
[128] B. Halliwell, Vitamin C: poison, prophylactic or panacea? Trends Biochem. Sci. 24 with advanced cancer. A controlled trial, N. Engl. J. Med. 301 (1979) 687–690.
(1999) 255–259. [160] C.G. Moertel, T.R. Fleming, E.T. Creagan, J. Rubin, M.J. O'Connell, M.M. Ames,
[129] B. Frei, S. Lawson, Vitamin C and cancer revisited, Proc. Natl. Acad. Sci. U. S. A. High-dose vitamin C versus placebo in the treatment of patients with advanced
105 (1998) 11037–11038. cancer who have had no prior chemotherapy. A randomized double-blind
[130] S. Rees, T.F. Slater, Ascorbic acid and lipid peroxidation: the cross-over effect, comparison, N. Engl. J. Med. 312 (1985) 137–141.
Acta Biochim. Biophys. Hung. 22 (1987) 241–249. [161] J.A. Drisko, J. Chapman, V.J. Hunter, The use of antioxidants with first-line che-
[131] G.R. Buettner, B.A. Jurkiewicz, Chemistry and biochemistry of ascorbic acid, In: motherapy in two cases of ovarian cancer, J. Am. Coll. Nutr. 22 (2003) 118–123.
in: E. Cadenas, L. Packer (Eds.), Handbook of Antioxidants, Marcel Dekker, [162] H.D. Riordan, J.J. Casciari, M.J. Gonzalez, N.H. Riordan, J.R. Miranda-Massari, P.
New York, 1996, pp. 91–115. Taylor, J.A. Jackson, A pilot clinical study of continuous intravenous ascorbate
[132] W. Wang, M.A. Knovich, L.G. Coffman, F.M. Torti, S.V. Torti, Serum ferritin: in terminal cancer patients, P. R. Health Sci. J. 24 (2005) 269–276.
past, present and future, Biochim. Biophys. Acta — Gen. Subj. 1800 (2010) [163] P. Sestili, G. Brandi, L. Brambilla, F. Cattabeni, O. Cantoni, Hydrogen peroxide
760–769. mediates the killing of U937 tumor cells elicited by pharmacologically attainable
[133] D.R. Richardson, P. Ponka, The molecular mechanisms of the metabolism and concentrations of ascorbic acid: cell death prevention by extracellular catalase
transport of iron in normal and neoplastic cells, Biochim. Biophys. Acta Rev. or catalase from cocultured erythrocytes or fibroblasts, J. Pharmacol. Exp. Ther.
Biomembr. 1331 (1997) 1–40. 277 (1996) 1719–1725.
[134] G.R. Buettner, The reaction of superoxide, formate radical, and hydrated electron [164] J. Verrax, P.B. Calderon, Pharmacologic concentrations of ascorbate are achieved
with transferrin and its model compound, Fe(III)-ethylenediamine-N,N′-bis by parenteral administration and exhibit antitumoral effects, Free Radic. Biol.
[2-(2-hydroxyphenyl) acetic acid] as studied by pulse radiolysis, J. Biol. Chem. Med. 47 (2009) 32–40.
262 (1987) 11995–11998. [165] M.V. Clement, J. Ramalingam, L.H. Long, B. Halliwell, The in vitro cytotoxicity
[135] S. Sirivech, E. Frieden, S. Osaki, The release of iron from horse spleen ferritin by of ascorbate depends on the culture medium used to perform the assay and
reduced flavins, Biochem. J. 143 (1974) 311–315. involves hydrogen peroxide, Antioxid. Redox Signal. 3 (2001) 157–163.
456 J. Du et al. / Biochimica et Biophysica Acta 1826 (2012) 443–457

[166] B.A. Wagner, L.M. Teesch, G.R. Buettner, B.E. Britigan, C.P. Burns, K.J. Reszka, superoxide dismutase mimic in RWPE-2 human prostate adenocarcinoma
Inactivation of anthracyclines by serum heme proteins, Chem. Res. Toxicol. 20 cells, Antioxid. Redox Signal. 6 (2004) 513–522.
(2007) 920–926. [198] J. Tian, D.M. Peehl, S.J. Knox, Metalloporphyrin synergizes with ascorbic acid to
[167] A.R. Giandomenico, G.E. Cerniglia, J.E. Biaglow, C.W. Stevens, C.J. Koch, The inhibit cancer cell growth through Fenton chemistry, Cancer Biother.
importance of sodium pyruvate in assessing damage produced by hydrogen Radiopharm. 25 (2010) 439–448.
peroxide, Free Radic. Biol. Med. 23 (1997) 426–434. [199] I. Batinic-Haberle, Z. Rajic, A. Tovmasyan, J.S. Reboucas, X. Ye, K.W. Leong, M.W.
[168] L.H. Long, B. Halliwell, Artefacts in cell culture: pyruvate as a scavenger of Dewhirst, Z. Vujaskovic, L. Benov, I. Spasojevic, Diverse functions of cationic
hydrogen peroxide generated by ascorbate or epigallocatechin gallate in cell Mn(III) N-substituted pyridylporphyrins, recognized as SOD mimics, Free
culture media, Biochem. Biophys. Res. Commun. 388 (2009) 700–704. Radic. Biol. Med. 51 (2011) 1035–1053.
[169] J.J. Casciari, N.H. Riordan, T.L. Schmidt, X.L. Meng, J.A. Jackson, H.D. Riordan, [200] X. Ye, D. Fels, A. Tovmasyan, K.M. Aird, C. Dedeugd, J.L. Allensworth, I. Kos, W.
Cytotoxicity of ascorbate, lipoic acid, and other antioxidants in hollow fibre in Park, I. Spasojevic, G.R. Devi, M.W. Dewhirst, K.W. Leong, I. Batinic-Haberle,
vitro tumours, Br. J. Cancer 84 (2001) 1544–1550. Cytotoxic effects of Mn(III) N-alkylpyridylporphyrins in the presence of cellular
[170] R. Radi, J.F. Turrens, L.Y. Chang, K.M. Bush, J.D. Crapo, B.A. Freeman, Detection of reductant, ascorbate, Free. Radic. Res. 45 (2011) 1289–1306.
catalase in rat heart mitochondria, J. Biol. Chem. 266 (1991) 22028–22034. [201] M. Klockars, T. Weber, P. Tanner, P.E. Hellstrom, T. Pettersson, Pleural fluid ferri-
[171] L. Benade, T. Howard, D. Burk, Synergistic killing of Ehrlich ascites carcinoma tin concentrations in human disease, J. Clin. Pathol. 38 (1985) 818–824.
cells by ascorbate and 3-amino-1,2,4,-triazole, Oncology 23 (1969) 33–43. [202] A. Weinberger, P. Simkin, Plasma proteins in synovial fluids of normal human
[172] G.F. Gaetani, S. Galiano, L. Canepa, A.M. Ferraris, H.N. Kirkman, Catalase and joints, Semin. Arthritis Rheum. 19 (1989) 66–76.
glutathione peroxidase are equally active in detoxification of hydrogen peroxide [203] B. Halliwell, Oxygen radicals, nitric oxide and human inflammatory joint dis-
in human erythrocytes, Blood 73 (1989) 334–339. ease, Ann. Rheum. Dis. 54 (1995) 505–510.
[173] G.F. Gaetani, H.N. Kirkman, R. Mangerini, A.M. Ferraris, Importance of catalase in [204] G. Baronzio, L. Schwartz, M. Kiselevsky, A. Guais, E. Sanders, G. Milanesi, M.
the disposal of hydrogen peroxide within human erythrocytes, Blood 84 (1994) Baronzio, I. Freitas, Tumor interstitial fluid as modulator of cancer inflammation,
325–330. thrombosis, immunity and angiogenesis, Anticancer. Res. 32 (2012) 405–414.
[174] C.F. Ng, F.Q. Schafer, G.R. Buettner, V.G.J. Rodgers, The rate of cellular hydrogen [205] J.A. Nagy, E.M. Masse, K.T. Herzberg, M.S. Meyers, K.-T. Yeo, T.-K. Yeo, T.M.
peroxide removal shows dependency on GSH: mathematical insight into in Sioussat, H.F. Dvorak, Pathogenesis of ascites tumor growth: vascular perme-
vivo H2O2 and GPx concentrations, Free. Radic. Res. 41 (2007) 1201–1211. ability factor, vascular hyperpermeability, and ascites fluid accumulation,
[175] K. Sasaki, S. Bannai, N. Makino, Kinetics of hydrogen peroxide elimination by Cancer Res. 55 (1995) 360–368.
human umbilical vein endothelial cells in culture, Biochim. Biophys. Acta Gen. [206] J.A. Nagy, K.T. Herzberg, J.M. Dvorak, H.F. Dvorak, Pathogenesis of malignant
Subj. 1380 (1998) 275–288. ascites formation: initiating events that lead to fluid accumulation, Cancer Res.
[176] F.M. Low, M.B. Hampton, C.C. Winterbourn, Peroxiredoxin 2 and peroxide 53 (1993) 2631–2643.
metabolism in the erythrocyte, Antioxid. Redox Signal. 10 (2008) 1621–1630. [207] M. Roche, P. Rondeau, N.R. Singh, E. Tarnus, E. Bourdon, The antioxidant proper-
[177] A.V. Peskin, F.M. Low, L.N. Paton, G.J. Maghzal, M.B. Hampton, C.C. ties of serum albumin, FEBS Lett. 582 (2008) 1783–1787.
Winterbourn, The high reactivity of peroxiredoxin 2 with H2O2 is not reflected [208] B. Halliwell, J.M.C. Gutteridge, The antioxidants of human extracellular fluids,
in its reaction with other oxidants and thiol reagents, J. Biol. Chem. 282 (2007) Arch. Biochem. Biophys. 280 (1990) 1–8.
11885–11892. [209] M.G. Espey, P. Chen, B. Chalmers, J. Drisko, A.Y. Sun, M. Levine, Q. Chen, Pharma-
[178] B. Chance, H. Sies, A. Boveris, Hydroperoxide metabolism in mammalian organs, cologic ascorbate synergizes with gemcitabine in preclinical models of pancreat-
Physiol. Rev. 59 (1979) 527–605. ic cancer, Free Radic. Biol. Med. 50 (2011) 1610–1619.
[179] F.Q. Schafer, G.R. Buettner, Redox environment of the cell as viewed through the [210] N.H. Riordan, H.D. Riordan, X. Meng, J.A. Jackson, Intravenous ascorbate as a
redox state of the glutathione disulfide/glutathione couple, Free Radic. Biol. tumor cytotoxic chemotherapeutic agent, Med. Hypotheses 44 (1995) 207–213.
Med. 30 (2001) 1191–1212. [211] H.D. Riordan, N.H. Riordan, J.A. Jackson, J.J. Casciari, R. Hunninghake, M.J. Gonzalez,
[180] J. Liu, M.M. Hinkhouse, W. Sun, C.J. Weydert, J.M. Ritchie, L.W. Oberley, J.J. Cullen, E.M. Mora, J.R. Miranda-Massari, N. Roscario, Intravenous vitamin C as a chemo-
Redox regulation of pancreatic cancer cell growth: role of glutathione peroxi- therapy agent: a report on clinical cases, P. R. Health Sci. J. 23 (2004) 115–118.
dase in the suppression of the malignant phenotype, Hum. Gene Ther. 15 [212] S.J. Padayatty, H.D. Riordan, S.M. Hewitt, A. Katz, L.J. Hoffer, M. Levine, Intrave-
(2004) 239–250. nously administered vitamin C as cancer therapy: three cases, CMAJ 174
[181] L.W. Oberley, Mechanism of the tumor suppressive effect of MnSOD (2006) 937–942.
overexpression, Biomed. Pharmacother. 59 (2005) 143–148. [213] S.J. Padayatty, A.Y. Sun, Q. Chen, M.G. Espey, J. Drisko, M. Levine, Vitamin C:
[182] H. Glickstein, R. Ben El, M. Shvartsman, Z.I. Cabantchik, Intracellular labile iron intravenous use by complementary and alternative medicine practitioners and
pools as direct targets of iron chelators: a fluorescence study of chelator action adverse effects, PLoS One 5 (2010) e11414.
in living cells, Blood 106 (2005) 3242–3250. [214] W.N. Tian, L.D. Braunstein, K. Apse, J. Pang, M. Rose, X. Tian, R.C. Stanton, Impor-
[183] P. Browne, O. Shalev, R.P. Hebbel, The molecular pathobiology of cell membrane tance of glucose-6-phosphate dehydrogenase activity in cell death, Am. J. Physiol.
iron: the sickle red cell as a model, Free Radic. Biol. Med. 24 (1998) 1040–1048. 276 (1999) C1121–C1131.
[184] S.L. Hempel, G.R. Buettner, D.A. Wessels, G.M. Galvan, Y.Q. O'Malley, Extracellu- [215] G.D. Campbell, M.H. Steinberg, J.D. Bower, Letter: Ascorbic acid-induced hemo-
lar Iron(II) can protect cells from hydrogen peroxide, Arch. Biochem. Biophys. lysis in G-6-PD deficiency, Ann. Intern. Med. 82 (1975) 810–815.
330 (1996) 401–408. [216] J.B. Mehta, S.B. Singhal, B.C. Mehta, Ascorbic-acid-induced haemolysis in G-6-PD
[185] F.Q. Schafer, S.Y. Qian, G.R. Buettner, Iron and free radical oxidations in cell deficiency, Lancet 336 (1990) 944.
membranes, Cell. Mol. Biol. (Noisy-le-grand) 46 (2000) 657–662. [217] D.C. Rees, H. Kelsey, J.D. Richards, Acute haemolysis induced by high dose ascorbic
[186] B.A. Jurkiewicz, G.R. Buettner, EPR detection of free radicals in UV-irradiated acid in glucose-6-phosphate dehydrogenase deficiency, BMJ 306 (1993) 841–842.
skin: mouse versus human, Photochem. Photobiol. 64 (1996). [218] K. Wong, C. Thomson, R.R. Bailey, S. McDiarmid, J. Gardner, Acute oxalate
[187] S.A. Bloomer, K.E. Brown, G.R. Buettner, K.C. Kregel, Dysregulation of hepatic nephropathy after a massive intravenous dose of vitamin C, Aust. N. Z. J. Med.
iron with aging: implications for heat stress-induced oxidative liver injury, 24 (1994) 410–411.
Am. J. Physiol. Regul. Integr. Comp. Physiol. 294 (2008) R1165–R1174. [219] A. Campbell, T. Jack, Acute reactions to mega ascorbic acid therapy in malignant
[188] K.T. Spencer, P.D. Lindower, G.R. Buettner, R.E. Kerber, Transition metal chela- disease, Scott. Med. J. 24 (1979) 151–153.
tors reduce directly measured myocardial free radical production during reper- [220] L.J. Hoffer, M. Levine, S. Assouline, D. Melnychuk, S.J. Padayatty, K. Rosadiule, C.
fusion, J. Cardiovas. Pharmacol. 32 (1998) 343–348. Rousseau, L. Robitaille, W.H.J. Miller, Phase I clinical trial of i.v. ascorbic acid in
[189] D.W. Reif, Ferritin as a source of iron for oxidative damage, Free Radic. Biol. Med. advanced malignancy, Ann. Oncol. 19 (2008) 1969–1974.
12 (1992) 417–427. [221] D.A. Monti, E. Mitchell, A.J. Bazzan, S. Littman, G. Zabrecky, C.J. Yeo, M.V. Pillai,
[190] T.C. Carmine, P. Evans, G. Bruchelt, R. Evans, R. Handgretinger, D. Niethammer, B. A.B. Newberg, S. Deshmukh, M. Levine, Phase I evaluation of intravenous
Halliwell, Presence of iron catalytic for free radical reactions in patients under- ascorbic acid in combination with gemcitabine and erlotinib in patients with
going chemotherapy: implications for therapeutic management, Cancer Lett. metastatic pancreatic cancer, PLoS One 7 (2012) e29794.
94 (1995) 219–226. [222] K.N. Prasad, P.K. Sinha, M. Ramanujam, A. Sakamoto, Sodium ascorbate potenti-
[191] H.L. Persson, Radiation-induced lysosomal iron reactivity: implications for ates the growth inhibitory effect of certain agents on neuroblastoma cells in cul-
radioprotective therapy, IUBMB Life 58 (2006) 395–401. ture, Proc. Natl. Acad. Sci. U. S. A. 76 (1979) 829–832.
[192] G.R. Buettner, Ascorbate autoxidation in the presence of iron and copper che- [223] C.M. Kurbacher, U. Wagner, B. Kolster, P.E. Andretti, D. Krebs, H.W. Bruckner,
lates, Free. Radic. Res. Commun. 1 (1986) 349–353. Ascorbic acid (vitamin C) improves the antineoplastic activity of doxorubicin,
[193] D.M. Miller, G.R. Buettner, S.D. Aust, Transition metals as catalysts of “autoxida- cisplatin, and paclitaxel in human breast carcinoma cells in vitro, Cancer Lett.
tion” reactions, Free Radic. Biol. Med. 8 (1990) 95–108. 103 (1996) 183–189.
[194] E. Kimoto, H. Tanaka, J. Gyotoku, F. Morishige, L. Pauling, Enhancement of [224] P.M. Herst, K.W.R. Broadley, J.L. Harper, M.J. McConnell, Pharmacological con-
antitumor activity of ascorbate against Ehrlich ascites tumor cells by the copper: centrations of ascorbate radiosensitize glioblastoma multiforme primary cells
glycylglycylhistidine complex, Cancer Res. 43 (1983) 824–828. by increasing oxidative DNA damage and inhibiting G2/M arrest, Free Radic.
[195] I. Batinić-Haberle, J.S. Rebouças, I. Spasojević, Superoxide dismutase mimics: Biol. Med. 52 (2012) 1486–1493.
chemistry, pharmacology, and therapeutic potential, Antioxid. Redox Signal. [225] C. Vollbracht, B. Schneider, V. Leendert, G. Weiss, L. Auerbach, J. Beuth, Intravenous
13 (2010) 877–918. vitamin C administration improves quality of life in breast cancer patients during
[196] P.R. Gardner, D.-D.H. Nguyen, C.W. White, Superoxide scavenging by Mn(II/III) chemo-/radiotherapy and aftercare: results of a retrospective, multicentre, epide-
tetrakis (1-Methyl-4-pyridyl) porphyrin in mammalian cells, Arch. Biochem. miological cohort study in Germany, In Vivo 25 (2011) 983–990.
Biophys. 325 (1996) 20–28. [226] M. Terpstra, K. Ugurbil, I. Tkac, Noninvasive quantification of human brain ascor-
[197] W. Zhong, T. Yan, M.M. Webber, T.D. Oberley, Alteration of cellular phenotype bate concentration using 1H NMR spectroscopy at 7 T, NMR Biomed. 23 (2010)
and responses to oxidative stress by manganese superoxide dismutase and a 227–232.
J. Du et al. / Biochimica et Biophysica Acta 1826 (2012) 443–457 457

[227] M. Terpstra, C. Torkelson, U. Emir, J.S. Hodges, S. Raatz, Noninvasive quantifica- [248] J.M. May, Z.-C. Qu, Ascorbate-dependent electron transfer across the human
tion of human brain antioxidant concentrations after an intravenous bolus of erythrocyte membrane, Biochim. Biophys. Acta Biomembr. 1421 (1999) 19–31.
vitamin C, NMR Biomed. 24 (2011) 521–528. [249] M. Han, S. Pendem, S.L. Teh, D.K. Sukumaran, F. Wu, J.X. Wilson, Ascorbate pro-
[228] S.T. Omaye, J.H. Skala, R.A. Jacob, Rebound effect with ascorbic acid in adult tects endothelial barrier function during septic insult: role of protein phospha-
males, Am. J. Clin. Nutr. 48 (1988) 379–380. tase type 2A, Free Radic. Biol. Med. 48 (2010) 128–135.
[229] C.S. Tsao, P.Y. Leung, Urinary ascorbic acid levels following the withdrawal of [250] J. May, L. Li, K. Hayslett, Z.-c. Qu, Ascorbate transport and recycling by
large doses of ascorbic acid in guinea pigs, J. Nutr. 118 (1988) 895–900. SH-SY5Y neuroblastoma cells: response to glutamate toxicity, Neurochem.
[230] J.D. Morrow, K.E. Hill, R.F. Burk, T.M. Nammour, K.F. Badr, L.J. Roberts, A series of Res. 31 (2006) 785–794.
prostaglandin F2-like compounds are produced in vivo in humans by a [251] T.L. Duarte, G.M. Almeida, G.D. Jones, Investigation of the role of extracellular
non-cyclooxygenase, free radical-catalyzed mechanism, Proc. Natl. Acad. Sci. H2O2 and transition metal ions in the genotoxic action of ascorbic acid in cell
U. S. A. 87 (1990) 9383–9387. culture models, Toxicol. Lett. 170 (2007) 57–65.
[231] G.L. Milne, S.C. Sanchez, E.S. Musiek, J.D. Morrow, Quantification of F2-isoprostanes [252] B. Peterkofsky, W. Prather, Cytotoxicity of ascorbate and other reducing agents
as a biomarker of oxidative stress, Nat. Protoc. 2 (2007) 221–226. towards cultured fibroblasts as a result of hydrogen peroxide formation, J. Cell.
[232] M.B. Kadiiska, B.C. Gladen, D.D. Baird, D. Germolec, L.B. Graham, C.E. Parker, A. Physiol. 90 (1977) 61–70.
Nyska, J.T. Wachsman, B.N. Ames, S. Basu, N. Brot, G.A. FitzGerald, R.A. Floyd, [253] N.A. Johnson, B.-H. Chen, S.-Y. Sung, C.-H. Liao, W.-C. Hsiao, L.W.K. Chung, C.-L.
M. George, J.W. Heinecke, G.E. Hatch, K. Hensley, J.A. Lawson, L.J. Marnett, J.D. Hsieh, A novel targeting modality for renal cell carcinoma: human osteocalcin
Morrow, D.M. Murray, J. Plastaras, L.J. Roberts II, J. Rokach, M.K. Shigenaga, R.S. promoter-mediated gene therapy synergistically induced by vitamin C and vita-
Sohal, J. Sun, R.R. Tice, D.H. Van Thiel, D. Wellner, P.B. Walter, K.B. Tomer, R.P. min D3, J. Gene Med. 12 (2010) 892–903.
Mason, J.C. Barrett, Biomarkers of oxidative stress study II: are oxidation prod- [254] C.H. Park, M. Amare, M.A. Savin, Growth suppression of human leukemic cells in
ucts of lipids, proteins, and DNA markers of CCl4 poisoning? Free Radic. Biol. vitro by L-ascorbic acid, Cancer Res. 40 (1980) 1062–1065.
Med. 38 (2005) 698–710. [255] J.M. Jamison, J. Gilloteaux, M.R. Nassiri, M. Venugopal, D.R. Neal, J.L. Summers, Cell
[233] M. Levine, S.J. Padayatty, M.G. Espey, Vitamin C: a concentration-function approach cycle arrest and autoschizis in a human bladder carcinoma cell line following vita-
yields pharmacology and therapeutic discoveries, Adv. Nutr. 2 (2011) 78–88. min C and vitamin K3 treatment, Biochem. Pharmacol. 67 (2004) 337–351.
[234] U.E. Emir, S. Raatz, S. McPherson, J.S. Hodges, C. Torkelson, P. Tawfik, T. White, M. [256] N. Rozanova, J.Z. Zhang, D.E. Heck, Catalytic therapy of cancer with porphyrins
Terpstra, Noninvasive quantification of ascorbate and glutathione concentration and ascorbate, Cancer Lett. 252 (2007) 216–224.
in the elderly human brain, NMR Biomed. 24 (2011) 888–894. [257] C. Berndt, T. Kurz, S. Bannenberg, R. Jacob, A. Holmgren, U.T. Brunk, Ascorbate
[235] K. Lönnrot, T. MetsÄ-KetelÄ, G. Molnár, J.-P. Ahonen, M. Latvala, J. Peltola, T. PietilÄ, and endocytosed Motexafin gadolinium induce lysosomal rupture, Cancer Lett.
H. Alho, The effect of ascorbate and ubiquinone supplementation on plasma and 307 (2011) 119–123.
CSF total antioxidant capacity, Free Radic. Biol. Med. 21 (1996) 211–217. [258] M.E. Solovieva, V.V. Soloviev, V.S. Akatov, Vitamin B12b increases the cytotoxic-
[236] H. Bayir, V.E. Kagan, Y.Y. Tyurina, V. Tyurin, R.A. Ruppel, P.D. Adelson, S.H. ity of short-time exposure to ascorbic acid, inducing oxidative burst and
Graham, K. Janesko, R.S. Clark, P.M. Kochanek, Assessment of antioxidant iron-dependent DNA damage, Euro. J. Phmacol. 566 (2007) 206–214.
reserves and oxidative stress in cerebrospinal fluid after severe traumatic [259] S. Biswas, X. Zhao, A.P. Mone, X. Mo, M. Vargo, D. Jarjoura, J.C. Byrd, N.
brain injury in infants and children, Pediatr. Res. 51 (2002) 571–578. Muthusamy, Arsenic trioxide and ascorbic acid demonstrate promising activity
[237] K. Voigt, A. Kontush, H.J. Stuerenburg, D. Muench-Harrach, H.C. Hansen, K. against primary human CLL cells in vitro, Leuk. Res. 34 (2010) 925–931.
Kunze, Decreased plasma and cerebrospinal fluid ascorbate levels in patients [260] E. Hahm, D.-H. Jin, J.S. Kang, Y.-I. Kim, S.-W. Hong, S.K. Lee, H.N. Kim, D.J. Jung, J.E.
with septic encephalopathy, Free. Radic. Res. 36 (2002) 735–739. Kim, D.H. Shin, Y.I. Hwang, Y.S. Kim, D.Y. Hur, Y. Yang, D. Cho, M.-S. Lee, W.J. Lee,
[238] F.E. Harrison, J.M. May, Vitamin C function in the brain: vital role of the ascor- The molecular mechanisms of vitamin C on cell cycle regulation in B16F10 mu-
bate transporter SVCT2, Free Radic. Biol. Med. 46 (2009) 719–730. rine melanoma, J. Cell. Biochem. 102 (2007) 1002–1010.
[239] M.C.M. Vissers, S.P. Gunningham, M.J. Morrison, G.U. Dachs, M.J. Currie, Modula- [261] P.D. Josephy, B. Palcic, L.D. Skarsgard, Ascorbate-enhanced cytotoxicity of
tion of hypoxia-inducible factor-1 alpha in cultured primary cells by intracellu- misonidazole, Nature 271 (1978) 370–372.
lar ascorbate, Free Radic. Biol. Med. 42 (2007) 765–772. [262] S. Belin, F. Kaya, G. Duisit, S. Giacometti, J. Ciccolini, M. Fontes, Antiproliferative
[240] W.S. Koh, W.H. Choi, S.J. Lee, C.S. Park, C.H. Park, Enhancement of plasmacytoma effect of ascorbic acid is associated with the inhibition of genes necessary to cell
cell growth by ascorbic acid is mediated via glucose 6-phosphate dehydroge- cycle progression, PLoS One 4 (2009) e4409.
nase, Cancer Res. Treat. 39 (2007) 22–29. [263] J. Verrax, J. Stockis, A. Tison, H.S. Taper, P.B. Calderon, Oxidative stress by
[241] V. Castranova, J.R. Wright, H.D. Colby, P.R. Miles, Ascorbate uptake by isolated rat ascorbate/menadione association kills K562 human chronic myelogenous leu-
alveolar macrophages and type II cells, J. Appl. Physiol. 54 (1983) 208–214. kaemia cells and inhibits its tumour growth in nude mice, Biochem. Pharmacol.
[242] J. Korcok, S.J. Dixon, T.C.Y. Lo, J.X. Wilson, Differential effects of glucose on 72 (2006) 671–680.
dehydroascorbic acid transport and intracellular ascorbate accumulation in [264] S. Ohtani, A. Iwamaru, W. Deng, K. Ueda, G. Wu, G. Jayachandran, S. Kondo,
astrocytes and skeletal myocytes, Brain Res. 993 (2003) 201–207. E.N. Atkinson, J.D. Minna, J.A. Roth, L. Ji, Tumor suppressor 101F6 and ascor-
[243] P. Bergsten, R. Yu, J. Kehrl, M. Levine, Ascorbic acid transport and distribution in bate synergistically and selectively inhibit non-small cell lung cancer growth
human B lymphocytes, Arch. Biochem. Biophys. 317 (1995) 208–214. by caspase-independent apoptosis and autophagy, Cancer Res. 67 (2007)
[244] J.M. May, Z.-C. Qu, Chelation of intracellular iron enhances endothelial barrier 6293–6303.
function: a role for vitamin C? Arch. Biochem. Biophys. 500 (2010) 162–168. [265] Y. Takemura, M. Satoh, K. Satoh, H. Hamada, Y. Sekido, S. Kubota, High dose of
[245] J.C. Reidling, V.S. Subramanian, T. Dahhan, M. Sadat, H.M. Said, Mechanisms and ascorbic acid induces cell death in mesothelioma cells, Biochem. Biophys. Res.
regulation of vitamin C uptake: studies of the hSVCT systems in human liver epi- Commun. 394 (2010) 249–253.
thelial cells, Am. J. Physiol. Gastrointest. Liver Physiol. 295 (2008) G1217–G1227. [266] C.-H. Yeom, G. Lee, J.H. Park, J. Yu, S.I. Park, S.Y. Yi, H.R. Lee, Y.S. Hong, J. Yang, S.
[246] G.G. Kramarenko, W.W. Wilke, D. Dayal, G.R. Buettner, F.Q. Schafer, Ascorbate Lee, High dose concentration administration of ascorbic acid inhibits tumor
enhances the toxicity of the photodynamic action of Verteporfin in HL-60 growth in BALB/C mice implanted with sarcoma 180 cancer cells via the restric-
cells, Free Radic. Biol. Med. 40 (2006) 1615–1627. tion of angiogenesis, J. Transl. Med. 7 (2009) 70–79.
[247] R.J. Steffner, L. Wu, A.C. Powers, J.M. May, Ascorbic acid recycling by cultured β [267] S. Park, E.S. Ahn, S. Lee, M. Jung, J.H. Park, S.Y. Yi, C.-H. Yeom, Proteomic analysis
cells: effects of increased glucose metabolism, Free Radic. Biol. Med. 37 (2004) reveals upregulation of RKIP in S-180 implanted BALB/C mouse after treatment
1612–1621. with ascorbic acid, J. Cell. Biochem. 106 (2009) 1136–1145.

Das könnte Ihnen auch gefallen